1
|
Thomas MJ, Xu H, Wang A, Sorci-Thomas MG. PCPE2: Expression of Multifunctional Extracellular Glycoprotein Associates With Diverse Cellular Functions. J Lipid Res 2024:100664. [PMID: 39374805 DOI: 10.1016/j.jlr.2024.100664] [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: 03/31/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024] Open
Abstract
Procollagen C-endopeptidase enhancer 2, known as PCPE2 or PCOC2 (gene name, PCOLCE2) is a glycoprotein that resides in the extracellular matrix, and is similar in domain organization to PCPE1 / PCPE, PCOC1 (PCOLCE1 /PCOLCE). Due to the many similarities between the two related proteins, PCPE2 has been assumed to have biological functions similar to PCPE. PCPE is a well-established enhancer of procollagen processing activating the enzyme, BMP-1. However, reports show that PCPE2 has a strikingly different tissue expression profile compared to PCPE. With that in mind and given the paucity of published studies on PCPE2, this review examines the current literature citing PCPE2 and its association with specific cell types and signaling pathways. Additionally, this review will present a brief history of PCPE2's discovery, highlighting structural and functional similarities and differences compared to PCPE. Considering the widespread use of RNA sequencing techniques to examine associations between cell-specific gene expression and disease states, we will show that PCPE2 is repeatedly found as a differentially regulated gene (DEG) significantly associated with a number of cellular processes, well beyond the scope of procollagen fibril processing.
Collapse
Affiliation(s)
- Michael J Thomas
- Department of Pharmacology & Toxicology, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Cardiovascular Research Center, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Hao Xu
- Department of Medicine, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Angela Wang
- Department of Medicine, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mary G Sorci-Thomas
- Department of Pharmacology & Toxicology, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Cardiovascular Research Center, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Medicine, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
2
|
Zein L, Grossmann J, Swoboda H, Borgel C, Wilke B, Awe S, Nist A, Stiewe T, Stehling O, Freibert SA, Adhikary T, Chung HR. Haptoglobin buffers lipopolysaccharides to delay activation of NFκB. Front Immunol 2024; 15:1401527. [PMID: 39416789 PMCID: PMC11479958 DOI: 10.3389/fimmu.2024.1401527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
It has remained yet unclear which soluble factors regulate the anti-inflammatory macrophage phenotype observed in both homeostasis and tumourigenesis. We show here that haptoglobin, a major serum protein with elusive immunoregulatory properties, binds and buffers bacterial lipopolysaccharides to attenuate activation of NFκB in macrophages. Haptoglobin binds different lipopolysaccharides with low micromolar affinities. Given its abundance, haptoglobin constitutes a buffer for serum-borne lipopolysaccharides, shielding them to safeguard against aberrant inflammatory reactions by reducing the amount of free lipopolysaccharides available for binding to TLR4. Concordantly, NFκB activation by haptoglobin-associated lipopolysaccharides was markedly delayed relative to stimulation with pure lipopolysaccharide. Our findings warrant evaluation of therapeutic benefits of haptoglobin for inflammatory conditions and re-evaluation of purification strategies. Finally, they allow to elucidate mechanisms of enhanced immunosuppression by oncofetal haptoglobin.
Collapse
Affiliation(s)
- Laura Zein
- Institute for Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University Marburg, Marburg, Germany
- Institute for Medical Bioinformatics and Biostatistics, Philipps University Marburg, Marburg, Germany
| | - Josina Grossmann
- Institute for Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University Marburg, Marburg, Germany
- Institute for Medical Bioinformatics and Biostatistics, Philipps University Marburg, Marburg, Germany
| | - Helena Swoboda
- Institute for Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University Marburg, Marburg, Germany
- Institute for Medical Bioinformatics and Biostatistics, Philipps University Marburg, Marburg, Germany
| | - Christina Borgel
- Institute for Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University Marburg, Marburg, Germany
- Institute for Medical Bioinformatics and Biostatistics, Philipps University Marburg, Marburg, Germany
| | - Bernhard Wilke
- Institute for Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University Marburg, Marburg, Germany
- Institute for Medical Bioinformatics and Biostatistics, Philipps University Marburg, Marburg, Germany
| | - Stephan Awe
- Institute for Molecular Biology and Tumor Research, Biomedical Research Center, Philipps University Marburg, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Center for Tumor Biology and Immunology, Philipps University Marburg, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Center for Tumor Biology and Immunology, Philipps University Marburg, Marburg, Germany
| | - Oliver Stehling
- Protein Biochemistry and Spectroscopy Core Facility, Center for Synthetic Microbiology, Philipps University Marburg, Marburg, Germany
- Institute of Cytobiology, Center for Synthetic Microbiology, Philipps University Marburg, Marburg, Germany
| | - Sven-Andreas Freibert
- Protein Biochemistry and Spectroscopy Core Facility, Center for Synthetic Microbiology, Philipps University Marburg, Marburg, Germany
- Institute of Cytobiology, Center for Synthetic Microbiology, Philipps University Marburg, Marburg, Germany
| | - Till Adhikary
- Institute for Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University Marburg, Marburg, Germany
- Institute for Medical Bioinformatics and Biostatistics, Philipps University Marburg, Marburg, Germany
| | - Ho-Ryun Chung
- Institute for Medical Bioinformatics and Biostatistics, Philipps University Marburg, Marburg, Germany
| |
Collapse
|
3
|
Napoli M, Bauer J, Bonod C, Goff SVL, Moali C. PCPE-2 (procollagen C-proteinase enhancer-2): the NON-IDENTICAL twin of PCPE-1. Matrix Biol 2024:S0945-053X(24)00113-6. [PMID: 39251075 DOI: 10.1016/j.matbio.2024.09.001] [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: 07/29/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
PCPE-2 was discovered at the beginning of this century, and was soon identified as a close homolog of PCPE-1 (procollagen C-proteinase enhancer 1). After the demonstration that it could also stimulate the proteolytic maturation of fibrillar procollagens by BMP-1/tolloid-like proteinases (BTPs), PCPE-2 did not attract much attention as it was thought to fulfill the same functions as PCPE-1 which was already well-described. However, the tissue distribution of PCPE-2 shows both common points and significant differences with PCPE-1, suggesting that their activities are not fully overlapping. Also, the recently established connections between PCPE-2 (gene name PCOLCE2) and several important diseases such as atherosclerosis, inflammatory diseases and cancer have highlighted the need for a thorough reappraisal of the in vivo roles of this regulatory protein. In this context, the recent finding that, while retaining the ability to bind fibrillar procollagens and to activate their C-terminal maturation, PCPE-2 can also bind BTPs and inhibit their activity has substantially extended its potential functions. In this review, we describe the current knowledge about PCPE-2 with a focus on collagen fibrillogenesis, lipid metabolism and inflammation, and discuss how we could further advance our understanding of PCPE-2-dependent biological processes.
Collapse
Affiliation(s)
- Manon Napoli
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Julien Bauer
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Christelle Bonod
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Sandrine Vadon-Le Goff
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Catherine Moali
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France.
| |
Collapse
|
4
|
Ammarah U, Pereira‐Nunes A, Delfini M, Mazzone M. From monocyte-derived macrophages to resident macrophages-how metabolism leads their way in cancer. Mol Oncol 2024; 18:1739-1758. [PMID: 38411356 PMCID: PMC11223613 DOI: 10.1002/1878-0261.13618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 02/28/2024] Open
Abstract
Macrophages are innate immune cells that play key roles during both homeostasis and disease. Depending on the microenvironmental cues sensed in different tissues, macrophages are known to acquire specific phenotypes and exhibit unique features that, ultimately, orchestrate tissue homeostasis, defense, and repair. Within the tumor microenvironment, macrophages are referred to as tumor-associated macrophages (TAMs) and constitute a heterogeneous population. Like their tissue resident counterpart, TAMs are plastic and can switch function and phenotype according to the niche-derived stimuli sensed. While changes in TAM phenotype are known to be accompanied by adaptive alterations in their cell metabolism, it is reported that metabolic reprogramming of macrophages can dictate their activation state and function. In line with these observations, recent research efforts have been focused on defining the metabolic traits of TAM subsets in different tumor malignancies and understanding their role in cancer progression and metastasis formation. This knowledge will pave the way to novel therapeutic strategies tailored to cancer subtype-specific metabolic landscapes. This review outlines the metabolic characteristics of distinct TAM subsets and their implications in tumorigenesis across multiple cancer types.
Collapse
Affiliation(s)
- Ummi Ammarah
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CentreUniversity of TorinoItaly
| | - Andreia Pereira‐Nunes
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
- Life and Health Sciences Research Institute (ICVS), School of MedicineUniversity of MinhoBragaPortugal
- ICVS/3B's‐PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | - Marcello Delfini
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
| |
Collapse
|
5
|
Wang Y, Sun Y, Li X, Yu X, Zhang K, Liu J, Tian Q, Zhang H, Du X, Wang S. Progress in the treatment of malignant ascites. Crit Rev Oncol Hematol 2024; 194:104237. [PMID: 38128628 DOI: 10.1016/j.critrevonc.2023.104237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 11/14/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Malignant ascites occurs as a symptom of the terminal stage of cancer, affecting the quality of life through abdominal distension, pain, nausea, anorexia, dyspnea and other symptoms. We describe the current main drug treatments in addition to surgery according to the traditional and new strategies. Traditional treatments were based on anti-tumor chemotherapy and traditional Chinese medicine treatments, as well as diuretics to relieve the patient's symptoms. New treatments mainly involve photothermal therapy, intestinal therapy and targeted immunity. This study emphasizes that both traditional and new therapies have certain advantages and disadvantages, and medication should be adjusted according to different periods of use and different patients. In conclusion, this article reviews the literature to systematically describe the primary treatment modalities for malignant ascites.
Collapse
Affiliation(s)
- Yiqiu Wang
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yunting Sun
- Hangzhou TCM Hospital Afflitiated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311121, China.
| | - Xinyue Li
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xiaoli Yu
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Keying Zhang
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jinglei Liu
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Qingchang Tian
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Honghua Zhang
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xiao Du
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Shuling Wang
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| |
Collapse
|
6
|
Geng Z, Pan X, Xu J, Jia X. Friend and foe: the regulation network of ascites components in ovarian cancer progression. J Cell Commun Signal 2023; 17:391-407. [PMID: 36227507 PMCID: PMC10409702 DOI: 10.1007/s12079-022-00698-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/11/2022] [Indexed: 10/17/2022] Open
Abstract
The tumor microenvironment (TME) and its complex role in cancer progression have been hotspots of cancer research in recent years. Ascites, which occurs frequently in patients with ovarian cancer especially in advanced stages, represents a unique TME. Malignant ascites contains abundant cellular and acellular components that play important roles in tumorigenesis, growth, metastasis, and chemoresistance of ovarian cancer through complex molecular mechanisms and signaling pathways. As a valuable liquid biopsy sample, ascites fluid is also of great significance for the prognostic analysis of ovarian cancer. The components of ovarian cancer ascites are generally considered to comprise tumor-promoting factors; however, in recent years studies have found that ascites also contains tumor-suppressing factors, raising new perspectives on interactions between ascites and tumors. Malignant ascites directly constitutes the ovarian cancer microenvironment, therefore, the study of its components will aid in the development of new therapeutic strategies. This article reviews the current research on tumor-promoting and tumor-suppressing factors and molecular mechanisms of their actions in ovarian cancer-derived ascites and therapeutic strategies targeting ascites, which may provide references for the development of novel therapeutic targets for ovarian cancer in the future.
Collapse
Affiliation(s)
- Zhe Geng
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Mochou Rd, Nanjing, 210004, China
| | - Xinxing Pan
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Mochou Rd, Nanjing, 210004, China
| | - Juan Xu
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Mochou Rd, Nanjing, 210004, China.
| | - Xuemei Jia
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Mochou Rd, Nanjing, 210004, China.
| |
Collapse
|
7
|
Zhu YT, Wu SY, Yang S, Ying J, Tian L, Xu HL, Zhang HP, Yao H, Zhang WY, Jin QQ, Yang YT, Jiang XY, Zhang N, Yao S, Zhou SG, Chen G. Identification and validation of a novel anoikis-related signature for predicting prognosis and immune landscape in ovarian serous cystadenocarcinoma. Heliyon 2023; 9:e18708. [PMID: 37554782 PMCID: PMC10404752 DOI: 10.1016/j.heliyon.2023.e18708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Ovarian serous cystadenocarcinoma (OSC) is the most prevalent histological subtype of ovarian cancer (OV) and presents a serious threat to women's health. Anoikis is an essential component of metastasis, and tumor cells can get beyond it to become viable. The impact of anoikis on OSC, however, has only been the topic of a few studies. METHODS The mRNA sequencing and clinical information of OSC came from The Cancer Genome Atlas Target Genotype-Tissue Expression (TCGA TARGET GTEx) dataset. Anoikis-related genes (ARGs) were collected by Harmonizome and GeneCards websites. Centered on these ARGs, we used unsupervised consensus clustering to explore potential tumor typing and filtered hub ARGs to create a model of predictive signature for OSC patients. Furthermore, we presented clinical specialists with a novel nomogram based on ARGs, revealing the underlying clinical relevance of this signature. Finally, we explored the immune microenvironment among various risk groups. RESULTS We identified 24 ARGs associated with the prognosis of OSC and classified OSC patients into three subtypes, and the subtype with the best prognosis was more enriched in immune-related pathways. Seven ARGs (ARHGEF7, NOTCH4, CASP2, SKP2, PAK4, LCK, CCDC80) were chosen to establish a risk model and a nomogram that can provide practical clinical decision support. Risk scores were found to be an independent and significant prognostic factor in OSC patients. The CIBERSORTx result revealed an inflammatory microenvironment is different for risk groups, and the proportion of immune infiltrates of Macrophages M1 is negatively correlated with risk score (rs = -0.21, P < 0.05). Ultimately, quantitative reverse transcription polymerase chain reaction (RT-PCR) was utilized to validate the expression of the seven pivotal ARGs. CONCLUSION In this study, based on seven ARGs, a risk model and nomogram established can be used for risk stratification and prediction of survival outcomes in patients with OSC, providing a reliable reference for individualized therapy of OSC patients.
Collapse
Affiliation(s)
- Yu-Ting Zhu
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Shuang-Yue Wu
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Song Yang
- Department of Pain Treatment, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Jie Ying
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Lu Tian
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Hong-Liang Xu
- Department of Pathology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - He-Ping Zhang
- Department of Pathology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Hui Yao
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Wei-Yu Zhang
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Qin-Qin Jin
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Yin-Ting Yang
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Xi-Ya Jiang
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Nan Zhang
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Shun Yao
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Shu-Guang Zhou
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Guo Chen
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| |
Collapse
|
8
|
Diray-Arce J, Fourati S, Doni Jayavelu N, Patel R, Maguire C, Chang AC, Dandekar R, Qi J, Lee BH, van Zalm P, Schroeder A, Chen E, Konstorum A, Brito A, Gygi JP, Kho A, Chen J, Pawar S, Gonzalez-Reiche AS, Hoch A, Milliren CE, Overton JA, Westendorf K, Cairns CB, Rouphael N, Bosinger SE, Kim-Schulze S, Krammer F, Rosen L, Grubaugh ND, van Bakel H, Wilson M, Rajan J, Steen H, Eckalbar W, Cotsapas C, Langelier CR, Levy O, Altman MC, Maecker H, Montgomery RR, Haddad EK, Sekaly RP, Esserman D, Ozonoff A, Becker PM, Augustine AD, Guan L, Peters B, Kleinstein SH. Multi-omic longitudinal study reveals immune correlates of clinical course among hospitalized COVID-19 patients. Cell Rep Med 2023; 4:101079. [PMID: 37327781 PMCID: PMC10203880 DOI: 10.1016/j.xcrm.2023.101079] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/31/2023] [Accepted: 05/16/2023] [Indexed: 06/18/2023]
Abstract
The IMPACC cohort, composed of >1,000 hospitalized COVID-19 participants, contains five illness trajectory groups (TGs) during acute infection (first 28 days), ranging from milder (TG1-3) to more severe disease course (TG4) and death (TG5). Here, we report deep immunophenotyping, profiling of >15,000 longitudinal blood and nasal samples from 540 participants of the IMPACC cohort, using 14 distinct assays. These unbiased analyses identify cellular and molecular signatures present within 72 h of hospital admission that distinguish moderate from severe and fatal COVID-19 disease. Importantly, cellular and molecular states also distinguish participants with more severe disease that recover or stabilize within 28 days from those that progress to fatal outcomes (TG4 vs. TG5). Furthermore, our longitudinal design reveals that these biologic states display distinct temporal patterns associated with clinical outcomes. Characterizing host immune responses in relation to heterogeneity in disease course may inform clinical prognosis and opportunities for intervention.
Collapse
Affiliation(s)
- Joann Diray-Arce
- Clinical and Data Coordinating Center, Boston Children's Hospital, Boston, MA 02115, USA; Precision Vaccines Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Slim Fourati
- Emory School of Medicine, Atlanta, GA 30322, USA
| | | | - Ravi Patel
- University of California San Francisco, San Francisco, CA 94115, USA
| | - Cole Maguire
- The University of Texas at Austin, Austin, TX 78712, USA
| | - Ana C Chang
- Clinical and Data Coordinating Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Ravi Dandekar
- University of California San Francisco, San Francisco, CA 94115, USA
| | - Jingjing Qi
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brian H Lee
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Patrick van Zalm
- Precision Vaccines Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew Schroeder
- University of California San Francisco, San Francisco, CA 94115, USA
| | - Ernie Chen
- Yale School of Medicine, New Haven, CT 06510, USA
| | | | | | | | - Alvin Kho
- Clinical and Data Coordinating Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jing Chen
- Clinical and Data Coordinating Center, Boston Children's Hospital, Boston, MA 02115, USA; Precision Vaccines Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Annmarie Hoch
- Clinical and Data Coordinating Center, Boston Children's Hospital, Boston, MA 02115, USA; Precision Vaccines Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Carly E Milliren
- Clinical and Data Coordinating Center, Boston Children's Hospital, Boston, MA 02115, USA
| | | | | | - Charles B Cairns
- Drexel University, Tower Health Hospital, Philadelphia, PA 19104, USA
| | | | | | | | - Florian Krammer
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lindsey Rosen
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD 20814, USA
| | | | - Harm van Bakel
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael Wilson
- University of California San Francisco, San Francisco, CA 94115, USA
| | - Jayant Rajan
- University of California San Francisco, San Francisco, CA 94115, USA
| | - Hanno Steen
- Precision Vaccines Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Walter Eckalbar
- University of California San Francisco, San Francisco, CA 94115, USA
| | - Chris Cotsapas
- Yale School of Medicine, New Haven, CT 06510, USA; Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| | | | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| | - Matthew C Altman
- Benaroya Research Institute, University of Washington, Seattle, WA 98101, USA
| | - Holden Maecker
- Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | | | - Elias K Haddad
- Drexel University, Tower Health Hospital, Philadelphia, PA 19104, USA
| | | | | | - Al Ozonoff
- Clinical and Data Coordinating Center, Boston Children's Hospital, Boston, MA 02115, USA; Precision Vaccines Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| | - Patrice M Becker
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD 20814, USA
| | - Alison D Augustine
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD 20814, USA
| | - Leying Guan
- Yale School of Public Health, New Haven, CT 06510, USA
| | - Bjoern Peters
- La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | | |
Collapse
|
9
|
El-Arabey AA, Alkhalil SS, Al-Shouli ST, Awadalla ME, Alhamdi HW, Almanaa TN, Mohamed SSEM, Abdalla M. Revisiting macrophages in ovarian cancer microenvironment: development, function and interaction. Med Oncol 2023; 40:142. [PMID: 37039909 PMCID: PMC10090027 DOI: 10.1007/s12032-023-01987-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/02/2023] [Indexed: 04/12/2023]
Abstract
Tumor-associated macrophages (TAMs) are an important component of the tumor microenvironment (TME) and have been linked to immunosuppression and poor prognosis. TAMs have been shown to be harmful in ovarian cancer (OC), with a positive correlation between their high levels of tumors and poor overall patient survival. These cells are crucial in the progression and chemoresistance of OC. The primary pro-tumoral role of TAMs is the release of cytokines, chemokines, enzymes, and exosomes that directly enhance the invasion potential and chemoresistance of OC by activating their pro-survival signalling pathways. TAMs play a crucial role in the metastasis of OC in the peritoneum and ascities by assisting in spheroid formation and cancer cell adhesion to the metastatic regions. Furthermore, TAMs interact with tumor protein p53 (TP53), exosomes, and other immune cells, such as stem cells and cancer-associated fibroblasts (CAFs) to support the progression and metastasis of OC. In this review we revisit development, functions and interactions of TAMs in the TME of OC patients to highlight and shed light on challenges and excitement down the road.
Collapse
Affiliation(s)
- Amr Ahmed El-Arabey
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, 11751 Egypt
| | - Samia S. Alkhalil
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Alquwayiyah, Riyadh, Saudi Arabia
| | - Samia T. Al-Shouli
- Immunology Unit, Pathology Department, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | | | - Heba W. Alhamdi
- Department of Biology, College of Sciences, King Khalid University, Abha, 61413 Saudi Arabia
| | - Taghreed N. Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Samah Saif Eldin M. Mohamed
- Department of Clinical Laboratory science, College of Applied Sciences, Shaqra University, Alquwayiyah, Riyadh, Saudi Arabia
| | - Mohnad Abdalla
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University, Jinan, 250022 Shandong China
| |
Collapse
|
10
|
Truxova I, Cibula D, Spisek R, Fucikova J. Targeting tumor-associated macrophages for successful immunotherapy of ovarian carcinoma. J Immunother Cancer 2023; 11:jitc-2022-005968. [PMID: 36822672 PMCID: PMC9950980 DOI: 10.1136/jitc-2022-005968] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is among the top five causes of cancer-related death in women, largely reflecting early, prediagnosis dissemination of malignant cells to the peritoneum. Despite improvements in medical therapies, particularly with the implementation of novel drugs targeting homologous recombination deficiency, the survival rates of patients with EOC remain low. Unlike other neoplasms, EOC remains relatively insensitive to immune checkpoint inhibitors, which is correlated with a tumor microenvironment (TME) characterized by poor infiltration by immune cells and active immunosuppression dominated by immune components with tumor-promoting properties, especially tumor-associated macrophages (TAMs). In recent years, TAMs have attracted interest as potential therapeutic targets by seeking to reverse the immunosuppression in the TME and enhance the clinical efficacy of immunotherapy. Here, we review the key biological features of TAMs that affect tumor progression and their relevance as potential targets for treating EOC. We especially focus on the therapies that might modulate the recruitment, polarization, survival, and functional properties of TAMs in the TME of EOC that can be harnessed to develop superior combinatorial regimens with immunotherapy for the clinical care of patients with EOC.
Collapse
Affiliation(s)
| | - David Cibula
- Gynecologic Oncology Center, Department of Obstetrics and Gynecology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Radek Spisek
- Sotio Biotech, Prague, Czech Republic,Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Jitka Fucikova
- Sotio Biotech, Prague, Czech Republic .,Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| |
Collapse
|
11
|
Ou C, Peng C, Wang Y, Lu S, Yu X, He Q, He A, Zhang L. CRTC2 promotes paclitaxel resistance by inducing autophagy in ovarian cancer in part via the PI3K-AKT signaling axis. J Cancer 2023; 14:1011-1023. [PMID: 37151390 PMCID: PMC10158520 DOI: 10.7150/jca.82233] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/16/2023] [Indexed: 05/09/2023] Open
Abstract
Background: Ovarian cancer is the most malignant gynecological disease, which seriously threatens female physical and mental health. Paclitaxel is a first-line chemotherapy drug in the clinical treatment of ovarian cancer, but drug resistance has become an important factor affecting the survival of ovarian cancer patients. However, the main mechanism of chemotherapy resistance in ovarian cancer remains unclear. In this study, we analyzed the Integrated Gene Expression Database (GEO) dataset using comprehensive bioinformatics tools to provide new therapeutic strategies and search for prognostic targets for ovarian cancer. Methods: Ovarian cancer related genes were extracted from GSE18520 by bioinformatics method. Differentially expressed genes (DEGs) were obtained by differential analysis, and related genes and functions were elucidated. The key gene CRTC2 was identified by prognostic analysis. Immunohistochemistry was used to detect the expression of CRTC2 in chemotherapy-resistant and chemotherapy-sensitive ovarian cancer tissues. Functional analysis (cell assay) confirmed the role of CRTC2 in paclitaxel resistance. Autophagy related proteins were detected by Western blot. Autophagy flux analysis was performed using the GFP/RFP-LC3 adenovirus reporter. Results: A total of 3,852 DEGs were identified in the GEO microarray dataset. Key genes were screened by prognostic analysis. We found that CRTC2 was highly expressed in chemoresistant tissues of ovarian cancer. In 110 patients with ovarian cancer, high expression of CRTC2 was associated with poorer prognostic factors and shorter survival. At the same time, we found that CRTC2 can promote the proliferation and invasion ability of ovarian cancer cells. In addition, CRTC2 can affect the expression of PI3K, AKT, autophagic flux and sensitivity to paclitaxel chemotherapy in ovarian cancer. Conclusion: CRTC2 can affect autophagy partially through PI3K-AKT signaling pathway, and then affect the sensitivity of ovarian cancer to paclitaxel chemotherapy. CRTC2 may be a potential predictor or target for ovarian cancer therapy.
Collapse
Affiliation(s)
- Chaoyang Ou
- Department of Gynecology Oncology, Nantong Tumor Hospital, The Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Chen Peng
- Department of Gynecology and Obstetrics, The Affiliated Hospital of Nantong University, Nantong, China
| | - Yilang Wang
- Department of Oncology, The Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, China
| | - Shiyu Lu
- Department of Gynecology Oncology, Nantong Tumor Hospital, The Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Xinli Yu
- Department of Gynecology Oncology, Nantong Tumor Hospital, The Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Qian He
- Department of Gynecology Oncology, Nantong Tumor Hospital, The Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Aiqin He
- Department of Gynecology Oncology, Nantong Tumor Hospital, The Affiliated Tumor Hospital of Nantong University, Nantong, China
- ✉ Corresponding authors: Li Zhang. E-mail: . Aiqin He. E-mail:
| | - Li Zhang
- Department of Cancer Research Center, Nantong Tumor Hospital, The Affiliated Tumor Hospital of Nantong University, Nantong, China
- ✉ Corresponding authors: Li Zhang. E-mail: . Aiqin He. E-mail:
| |
Collapse
|
12
|
Liu T, Li Y, Wang X, Yang X, Fu Y, Zheng Y, Gong H, He Z. The role of interferons in ovarian cancer progression: Hinderer or promoter? Front Immunol 2022; 13:1087620. [PMID: 36618371 PMCID: PMC9810991 DOI: 10.3389/fimmu.2022.1087620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer (OC) is a common gynecologic malignancy with poor prognosis and high mortality. Changes in the OC microenvironment are closely related to the genesis, invasion, metastasis, recurrence, and drug-resistance. The OC microenvironment is regulated by Interferons (IFNs) known as a type of important cytokines. IFNs have a bidirectional regulation for OC cells growth and survival. Meanwhile, IFNs positively regulate the recruitment, differentiation and activation of immune cells. This review summarizes the secretion and the role of IFNs. In particular, we mainly elucidate the actions played by IFNs in various types of therapy. IFNs assist radiotherapy, targeted therapy, immunotherapy and biotherapy for OC, except for some IFN pathways that may cause chemo-resistance. In addition, we present some advances in OC treatment with the help of IFN pathways. IFNs have the ability to powerfully modulate the tumor microenvironment and can potentially provide new combination strategies for OC treatment.
Collapse
Affiliation(s)
- Taiqing Liu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yinqi Li
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyu Wang
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaodong Yang
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yunhai Fu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yeteng Zheng
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hanlin Gong
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Hanlin Gong, ; Zhiyao He,
| | - Zhiyao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China,Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China,*Correspondence: Hanlin Gong, ; Zhiyao He,
| |
Collapse
|
13
|
Sommerfeld L, Knuth I, Finkernagel F, Pesek J, Nockher WA, Jansen JM, Wagner U, Nist A, Stiewe T, Müller-Brüsselbach S, Müller R, Reinartz S. Prostacyclin Released by Cancer-Associated Fibroblasts Promotes Immunosuppressive and Pro-Metastatic Macrophage Polarization in the Ovarian Cancer Microenvironment. Cancers (Basel) 2022; 14:cancers14246154. [PMID: 36551640 PMCID: PMC9776493 DOI: 10.3390/cancers14246154] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
Metastasis of high-grade ovarian carcinoma (HGSC) is orchestrated by soluble mediators of the tumor microenvironment. Here, we have used transcriptomic profiling to identify lipid-mediated signaling pathways encompassing 41 ligand-synthesizing enzymes and 23 cognate receptors in tumor, immune and stroma cells from HGSC metastases and ascites. Due to its strong association with a poor clinical outcome, prostacyclin (PGI2) synthase (PTGIS) is of particular interest in this signaling network. PTGIS is highly expressed by cancer-associated fibroblasts (CAF), concomitant with elevated PGI2 synthesis, whereas tumor-associated macrophages (TAM) exhibit the highest expression of its surface receptor (PTGIR). PTGIR activation by PGI2 agonists triggered cAMP accumulation and induced a mixed-polarization macrophage phenotype with altered inflammatory gene expression, including CXCL10 and IL12A repression, as well as reduced phagocytic capability. Co-culture experiments provided further evidence for the interaction of CAF with macrophages via PGI2, as the effect of PGI2 agonists on phagocytosis was mitigated by cyclooxygenase inhibitors. Furthermore, conditioned medium from PGI2-agonist-treated TAM promoted tumor adhesion to mesothelial cells and migration in a PTGIR-dependent manner, and PTGIR activation induced the expression of metastasis-associated and pro-angiogenic genes. Taken together, our study identifies a PGI2/PTGIR-driven crosstalk between CAF, TAM and tumor cells, promoting immune suppression and a pro-metastatic environment.
Collapse
Affiliation(s)
- Leah Sommerfeld
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Isabel Knuth
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Florian Finkernagel
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
- Bioinformatics Spectrometry Core Facility, Philipps University, 35043 Marburg, Germany
| | - Jelena Pesek
- Medical Mass Spectrometry Core Facility, Philipps University, 35043 Marburg, Germany
| | - Wolfgang A. Nockher
- Medical Mass Spectrometry Core Facility, Philipps University, 35043 Marburg, Germany
| | - Julia M. Jansen
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital (UKGM), 35043 Marburg, Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital (UKGM), 35043 Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Sabine Müller-Brüsselbach
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Rolf Müller
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
- Correspondence: (R.M.); (S.R.)
| | - Silke Reinartz
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
- Correspondence: (R.M.); (S.R.)
| |
Collapse
|
14
|
Fanale D, Dimino A, Pedone E, Brando C, Corsini LR, Filorizzo C, Fiorino A, Lisanti MC, Magrin L, Randazzo U, Bazan Russo TD, Russo A, Bazan V. Prognostic and Predictive Role of Tumor-Infiltrating Lymphocytes (TILs) in Ovarian Cancer. Cancers (Basel) 2022; 14:4344. [PMID: 36139508 PMCID: PMC9497073 DOI: 10.3390/cancers14184344] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 12/12/2022] Open
Abstract
In the last decade, tumor-infiltrating lymphocytes (TILs) have been recognized as clinically relevant prognostic markers for improved survival, providing the immunological basis for the development of new therapeutic strategies and showing a significant prognostic and predictive role in several malignancies, including ovarian cancer (OC). In fact, many OCs show TILs whose typology and degree of infiltration have been shown to be strongly correlated with prognosis and survival. The OC histological subtype with the higher presence of TILs is the high-grade serous carcinoma (HGSC) followed by the endometrioid subtype, whereas mucinous and clear cell OCs seem to contain a lower percentage of TILs. The abundant presence of TILs in OC suggests an immunogenic potential for this tumor. Despite the high immunogenic potential, OC has been described as a highly immunosuppressive tumor with a high expression of PD1 by TILs. Although further studies are needed to better define their role in prognostic stratification and the therapeutic implication, intraepithelial TILs represent a relevant prognostic factor to take into account in OC. In this review, we will discuss the promising role of TILs as markers which are able to reflect the anticancer immune response, describing their potential capability to predict prognosis and therapy response in OC.
Collapse
Affiliation(s)
- Daniele Fanale
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Alessandra Dimino
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Erika Pedone
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Chiara Brando
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Lidia Rita Corsini
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Clarissa Filorizzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Alessia Fiorino
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Maria Chiara Lisanti
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Luigi Magrin
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Ugo Randazzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Tancredi Didier Bazan Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
15
|
Zhang Y, Ouyang D, Chen YH, Xia H. Peritoneal resident macrophages in tumor metastasis and immunotherapy. Front Cell Dev Biol 2022; 10:948952. [PMID: 36035994 PMCID: PMC9402905 DOI: 10.3389/fcell.2022.948952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022] Open
Abstract
Macrophages residing in various tissues play crucial roles in innate immunity, tissue repair, and immune homeostasis. The development and differentiation of macrophages in non-lymphoid tissues are highly regulated by the tissue microenvironment. Peritoneum provides a unique metastatic niche for certain types of tumor cells. As the dominant immune cell type in peritoneal cavity, macrophages control the immune response to tumor and influence the efficacy of anti-tumor therapy. Considering the heterogeneity of macrophages in origin, metabolism, and function, it is always challenging to define the precise roles of macrophages in tumor microenvironment. We review here recent progresses in peritoneal resident macrophage research in the context of physiological and metastatic tumor conditions, which may benefit the development of new anti-tumor therapies through targeting macrophages.
Collapse
Affiliation(s)
- Yu Zhang
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dongyun Ouyang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Youhai H. Chen
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Houjun Xia
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- *Correspondence: Houjun Xia,
| |
Collapse
|
16
|
Spheroid Formation and Peritoneal Metastasis in Ovarian Cancer: The Role of Stromal and Immune Components. Int J Mol Sci 2022; 23:ijms23116215. [PMID: 35682890 PMCID: PMC9181487 DOI: 10.3390/ijms23116215] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer (OC) is one of the most common gynecological cancers, with the worst prognosis and the highest mortality rate. Peritoneal dissemination (or carcinomatosis) accompanied by ascites formation is the most unfavorable factor in the progression and recurrence of OC. Tumor cells in ascites are present as either separate cells or, more often, as cell aggregates, i.e., spheroids which promote implantation on the surface of nearby organs and, at later stages, metastases to distant organs. Malignant ascites comprises a unique tumor microenvironment; this fact may be of relevance in the search for new prognostic and predictive factors that would make it possible to personalize the treatment of patients with OC. However, the precise mechanisms of spheroid formation and carcinomatosis are still under investigation. Here, we summarize data on ascites composition as well as the activity of fibroblasts and macrophages, the key stromal and immune components, in OC ascites. We describe current knowledge about the role of fibroblasts and macrophages in tumor spheroid formation, and discuss the specific functions of fibroblasts, macrophages and T cells in tumor peritoneal dissemination and implantation.
Collapse
|
17
|
Hammoud MK, Dietze R, Pesek J, Finkernagel F, Unger A, Bieringer T, Nist A, Stiewe T, Bhagwat AM, Nockher WA, Reinartz S, Müller-Brüsselbach S, Graumann J, Müller R. Arachidonic acid, a clinically adverse mediator in the ovarian cancer microenvironment, impairs JAK-STAT signaling in macrophages by perturbing lipid raft structures. Mol Oncol 2022; 16:3146-3166. [PMID: 35451191 PMCID: PMC9441005 DOI: 10.1002/1878-0261.13221] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/29/2022] [Accepted: 04/20/2022] [Indexed: 11/08/2022] Open
Abstract
Survival of ovarian carcinoma is associated with the abundance of immunosuppressed CD163highCD206high tumor‐associated macrophages (TAMs) and high levels of arachidonic acid (AA) in the tumor microenvironment. Here, we show that both associations are functionally linked. Transcriptional profiling revealed that high CD163 and CD206/MRC1 expression in TAMs is strongly associated with an inhibition of cytokine‐triggered signaling, mirrored by an impaired transcriptional response to interferons and IL‐6 in monocyte‐derived macrophages by AA. This inhibition of pro‐inflammatory signaling is caused by dysfunctions of the cognate receptors, indicated by the inhibition of JAK1, JAK2, STAT1, and STAT3 phosphorylation, and by the displacement of the interferon receptor IFNAR1, STAT1 and other immune‐regulatory proteins from lipid rafts. AA exposure led to a dramatic accumulation of free AA in lipid rafts, which appears to be mechanistically crucial, as the inhibition of its incorporation into phospholipids did not affect the AA‐mediated interference with STAT1 phosphorylation. Inhibition of interferon‐triggered STAT1 phosphorylation by AA was reversed by water‐soluble cholesterol, known to prevent the perturbation of lipid raft structure by AA. These findings suggest that the pharmacologic restoration of lipid raft functions in TAMs may contribute to the development new therapeutic approaches.
Collapse
Affiliation(s)
- Mohamad K Hammoud
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Raimund Dietze
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Jelena Pesek
- Medical Mass Spectrometry Core Facility, Philipps University, Marburg, Germany
| | - Florian Finkernagel
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Annika Unger
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Tim Bieringer
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany.,Hochschule Landshut, 84036, Landshut, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Aditya M Bhagwat
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany.,The German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - W Andreas Nockher
- Medical Mass Spectrometry Core Facility, Philipps University, Marburg, Germany
| | - Silke Reinartz
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | | | - Johannes Graumann
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany.,The German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Institute for Translational Proteomics, Philipps University, Marburg, Germany
| | - Rolf Müller
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| |
Collapse
|
18
|
Chen Q, Cai L, Liang J. Construction of prognosis model of bladder cancer based on transcriptome. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:79-86. [PMID: 35462469 PMCID: PMC9109759 DOI: 10.3724/zdxbyxb-2021-0368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/18/2022] [Indexed: 06/14/2023]
Abstract
OBJECTIVE To screen for prognosis related genes in bladder cancer, and to establish prognosis model of bladder cancer. METHODS The clinical information and bladder tissue RNA sequencing data of 406 bladder cancer patients, and the bladder tissue RNA sequencing data of 28 healthy individuals were downloaded from The Cancer Genome Atlas (TCGA) database, Genotype-Tissue Expression (GTEx) database through the UCSC Xena platform. The weighted gene co-expression network analysis (WGCNA), univariate Cox regression, LASSO regression analysis and multivariate Cox regression analysis were used to screen the prognosis-related genes of bladder cancer and the prognostic model was established. The prognostic model was evaluated with receiver operator characteristic curve (ROC curve). RESULTS A total of 2308 differentially expressed genes related to bladder cancer were obtained from the analysis. Six gene modules were obtained by WGCNA, and 829 genes with significant effect on bladder cancer prognosis were screened out. Univariate Cox regression and LASSO regression analysis showed that 24 genes were related to the prognosis of bladder cancer patients. Multivariate Cox regression analysis revealed 9 genes as independent predictors in training set, namely ADCY9, MAFG_DT, EMP1, CAST, PCOLCE2, LTBP1, CSPG4, NXPH4, SLC1A6, which were used to establish the prognosis model of bladder cancer patients. The 3-year survival rates of the high-risk group and the low-risk group in the training set were 31.814% and 59.821%, respectively. The 3-year survival rates of the high-risk group and the low-risk group in the test set were 32.745% and 68.932%, respectively. The areas under the ROC curve of the model for predicting the prognosis of bladder cancer patients in both the training set and the test set were above 0.7. CONCLUSION The established model in this study has good predictive ability for the survival of bladder cancer patients.
Collapse
Affiliation(s)
- Qiu Chen
- 1. Yangzhou University Medical College, Yangzhou 225001, Jiangsu Province, China
| | - Liangliang Cai
- 1. Yangzhou University Medical College, Yangzhou 225001, Jiangsu Province, China
- 2. Institute of Translational Medicine, Yangzhou University, Yangzhou 225001, Jiangsu Province, China
- 3. Jiangsu Provincial Key Laboratory of Geriatric Disease Prevention and Control, Yangzhou 225001, Jiangsu Province, China
| | - Jingyan Liang
- 1. Yangzhou University Medical College, Yangzhou 225001, Jiangsu Province, China
- 2. Institute of Translational Medicine, Yangzhou University, Yangzhou 225001, Jiangsu Province, China
- 3. Jiangsu Provincial Key Laboratory of Geriatric Disease Prevention and Control, Yangzhou 225001, Jiangsu Province, China
| |
Collapse
|
19
|
Constructing a thyroid cancer prognostic risk model based on CD8 + T cell associated genes. Cent Eur J Immunol 2022; 47:234-245. [PMID: 36817266 PMCID: PMC9896991 DOI: 10.5114/ceji.2022.119171] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/18/2022] [Indexed: 11/17/2022] Open
Abstract
Thyroid cancer (TC) is a common and curable endocrine tumor occurring in the head and neck characterized by a low mortality rate compared to other malignancies. In this study, the immune microenvironment of TC was investigated to identify biomarkers. The mRNA and clinical data available in this study were accessed from The Cancer Genome Atlas-Thyroid Cancer (TCGA-THCA) dataset. Differences in immune infiltration levels of TC and normal samples were assessed by CIBERSORT. Thyroid cancer samples were classified into high- and low-abundance groups according to the median abundance of immune cell infiltration, and CD8+ T cells were notably correlated with the survival status. Differential expression analysis was conducted on CD8+ T cells to obtain immune-related differentially expressed genes (DEGs). Subsequently, a prognostic risk model was established through Cox regression analysis. According to the median risk score, samples in the training set and validation set were assigned to high- and low-risk groups. The survival and ROC curves demonstrated that the model possesses favorable prognostic prediction ability. Furthermore, the results of gene set enrichment analysis (GSEA) indicated differences between the high- and low-risk groups in terms of ECM receptor interaction and transforming growth factor β (TGF-β) signaling pathways. The tumor microenvironment of TC samples was evaluated by ESTIMATE, which showed that stromal scores were higher in the high-risk group. Finally, simple-sample GSEA (ssGSEA) was performed on TC samples. The results indicated a higher infiltration level of NK cells in the low-risk group, as well as a lower level in the high-risk group. In terms of immune function-related gene sets, genes related to APC co-inhibition, cytolytic activity, HLA and T cell co-inhibition were observed to present higher expression levels in the low-risk group. In general, this study built a 6-gene prognostic risk assessment model based on CD8+ T cells through bioinformatics analysis, which is expected to be a reference for clinicians to judge the prognosis of TC patients.
Collapse
|
20
|
Menga A, Favia M, Spera I, Vegliante MC, Gissi R, De Grassi A, Laera L, Campanella A, Gerbino A, Carrà G, Canton M, Loizzi V, Pierri CL, Cormio G, Mazzone M, Castegna A. N-acetylaspartate release by glutaminolytic ovarian cancer cells sustains protumoral macrophages. EMBO Rep 2021; 22:e51981. [PMID: 34260142 PMCID: PMC8419692 DOI: 10.15252/embr.202051981] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 06/10/2021] [Accepted: 06/21/2021] [Indexed: 02/01/2023] Open
Abstract
Glutaminolysis is known to correlate with ovarian cancer aggressiveness and invasion. However, how this affects the tumor microenvironment is elusive. Here, we show that ovarian cancer cells become addicted to extracellular glutamine when silenced for glutamine synthetase (GS), similar to naturally occurring GS-low, glutaminolysis-high ovarian cancer cells. Glutamine addiction elicits a crosstalk mechanism whereby cancer cells release N-acetylaspartate (NAA) which, through the inhibition of the NMDA receptor, and synergistically with IL-10, enforces GS expression in macrophages. In turn, GS-high macrophages acquire M2-like, tumorigenic features. Supporting this in␣vitro model, in silico data and the analysis of ascitic fluid isolated from ovarian cancer patients prove that an M2-like macrophage phenotype, IL-10 release, and NAA levels positively correlate with disease stage. Our study uncovers the unprecedented role of glutamine metabolism in modulating macrophage polarization in highly invasive ovarian cancer and highlights the anti-inflammatory, protumoral function of NAA.
Collapse
Affiliation(s)
- Alessio Menga
- Department of Molecular Biotechnologies and Health SciencesUniversity of TurinTurinItaly
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
- Molecular Biotechnology CenterTurinItaly
| | - Maria Favia
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Iolanda Spera
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
| | - Maria C Vegliante
- Haematology and Cell Therapy UnitIRCCS‐Istituto Tumori ‘Giovanni Paolo II'BariItaly
| | - Rosanna Gissi
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
| | - Anna De Grassi
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
| | - Luna Laera
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
| | - Annalisa Campanella
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
| | - Andrea Gerbino
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
| | - Giovanna Carrà
- Molecular Biotechnology CenterTurinItaly
- Department of Clinical and Biological SciencesUniversity of TurinOrbassanoItaly
| | - Marcella Canton
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza ‐ IRPPadovaItaly
| | - Vera Loizzi
- Policlinico University of Bari “Aldo Moro”BariItaly
| | - Ciro L Pierri
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
| | - Gennaro Cormio
- Policlinico University of Bari “Aldo Moro”BariItaly
- Gynecologic Oncology UnitIRCCSIstituto Tumori Giovanni Paolo IIBariItaly
| | - Massimiliano Mazzone
- Department of Molecular Biotechnologies and Health SciencesUniversity of TurinTurinItaly
- Molecular Biotechnology CenterTurinItaly
- Laboratory of Tumor Inflammation and AngiogenesisCenter for Cancer BiologyDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Alessandra Castegna
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza ‐ IRPPadovaItaly
| |
Collapse
|
21
|
Rickard BP, Conrad C, Sorrin AJ, Ruhi MK, Reader JC, Huang SA, Franco W, Scarcelli G, Polacheck WJ, Roque DM, del Carmen MG, Huang HC, Demirci U, Rizvi I. Malignant Ascites in Ovarian Cancer: Cellular, Acellular, and Biophysical Determinants of Molecular Characteristics and Therapy Response. Cancers (Basel) 2021; 13:4318. [PMID: 34503128 PMCID: PMC8430600 DOI: 10.3390/cancers13174318] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/17/2021] [Accepted: 08/22/2021] [Indexed: 12/27/2022] Open
Abstract
Ascites refers to the abnormal accumulation of fluid in the peritoneum resulting from an underlying pathology, such as metastatic cancer. Among all cancers, advanced-stage epithelial ovarian cancer is most frequently associated with the production of malignant ascites and is the leading cause of death from gynecologic malignancies. Despite decades of evidence showing that the accumulation of peritoneal fluid portends the poorest outcomes for cancer patients, the role of malignant ascites in promoting metastasis and therapy resistance remains poorly understood. This review summarizes the current understanding of malignant ascites, with a focus on ovarian cancer. The first section provides an overview of heterogeneity in ovarian cancer and the pathophysiology of malignant ascites. Next, analytical methods used to characterize the cellular and acellular components of malignant ascites, as well the role of these components in modulating cell biology, are discussed. The review then provides a perspective on the pressures and forces that tumors are subjected to in the presence of malignant ascites and the impact of physical stress on therapy resistance. Treatment options for malignant ascites, including surgical, pharmacological and photochemical interventions are then discussed to highlight challenges and opportunities at the interface of drug discovery, device development and physical sciences in oncology.
Collapse
Affiliation(s)
- Brittany P. Rickard
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, and North Carolina State University, Raleigh, NC 27599, USA; (M.K.R.); (S.A.H.); (W.J.P.)
| | - Christina Conrad
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (C.C.); (A.J.S.); (G.S.); (H.-C.H.)
| | - Aaron J. Sorrin
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (C.C.); (A.J.S.); (G.S.); (H.-C.H.)
| | - Mustafa Kemal Ruhi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, and North Carolina State University, Raleigh, NC 27599, USA; (M.K.R.); (S.A.H.); (W.J.P.)
| | - Jocelyn C. Reader
- Department of Obstetrics, Gynecology and Reproductive Medicine, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (J.C.R.); (D.M.R.)
- Marlene and Stewart Greenebaum Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Stephanie A. Huang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, and North Carolina State University, Raleigh, NC 27599, USA; (M.K.R.); (S.A.H.); (W.J.P.)
| | - Walfre Franco
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, MA 01854, USA;
| | - Giuliano Scarcelli
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (C.C.); (A.J.S.); (G.S.); (H.-C.H.)
| | - William J. Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, and North Carolina State University, Raleigh, NC 27599, USA; (M.K.R.); (S.A.H.); (W.J.P.)
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dana M. Roque
- Department of Obstetrics, Gynecology and Reproductive Medicine, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (J.C.R.); (D.M.R.)
- Marlene and Stewart Greenebaum Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Marcela G. del Carmen
- Division of Gynecologic Oncology, Vincent Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (C.C.); (A.J.S.); (G.S.); (H.-C.H.)
- Marlene and Stewart Greenebaum Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA;
| | - Imran Rizvi
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, and North Carolina State University, Raleigh, NC 27599, USA; (M.K.R.); (S.A.H.); (W.J.P.)
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
22
|
Dai YH, Wang YF, Shen PC, Lo CH, Yang JF, Lin CS, Chao HL, Huang WY. Radiosensitivity index emerges as a potential biomarker for combined radiotherapy and immunotherapy. NPJ Genom Med 2021; 6:40. [PMID: 34078917 PMCID: PMC8172905 DOI: 10.1038/s41525-021-00200-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
In the era of immunotherapy, there lacks of a reliable genomic predictor to identify optimal patient populations in combined radiotherapy and immunotherapy (CRI). The purpose of this study is to investigate whether genomic scores defining radiosensitivity are associated with immune response. Genomic data from Merged Microarray-Acquired dataset (MMD) were established and the Cancer Genome Atlas (TCGA) were obtained. Based on rank-based regression model including 10 genes, radiosensitivity index (RSI) was calculated. A total of 12832 primary tumours across 11 major cancer types were analysed for the association with DNA repair, cellular stemness, macrophage polarisation, and immune subtypes. Additional 585 metastatic tissues were extracted from MET500. RSI was stratified into RSI-Low and RSI-High by a cutpoint of 0.46. Proteomic differential analysis was used to identify significant proteins according to RSI categories. Gene Set Variance Analysis (GSVA) was applied to measure the genomic pathway activity (18 genes for T-cell inflamed activity). Kaplan-Meier analysis was performed for survival analysis. RSI was significantly associated with homologous DNA repair, cancer stemness and immune-related molecular features. Lower RSI was associated with higher fraction of M1 macrophage. Differential proteomic analysis identified significantly higher TAP2 expression in RSI-Low colorectal tumours. In the TCGA cohort, dominant interferon-γ (IFN-γ) response was characterised by low RSI and predicted better response to programmed cell death 1 (PD-1) blockade. In conclusion, in addition to radiation response, our study identified RSI to be associated with various immune-related features and predicted response to PD-1 blockade, thus, highlighting its potential as a candidate biomarker for CRI.
Collapse
Affiliation(s)
- Yang-Hong Dai
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ying-Fu Wang
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Po-Chien Shen
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Hsiang Lo
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jen-Fu Yang
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Shu Lin
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsing-Lung Chao
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Radiation Oncology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Wen-Yen Huang
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan. .,Instititue of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
23
|
Serum Levels of S100A11 and MMP-9 in Patients with Epithelial Ovarian Cancer and Their Clinical Significance. BIOMED RESEARCH INTERNATIONAL 2021; 2021:7341247. [PMID: 33763485 PMCID: PMC7952149 DOI: 10.1155/2021/7341247] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 12/29/2020] [Accepted: 01/07/2021] [Indexed: 11/17/2022]
Abstract
Objective To investigate the serum levels of calgizzarin (S100A11) and matrix metalloproteinase-9 (MMP9) in patients with epithelial ovarian cancer (EOC) and determine their clinical significance. Methods Serum levels of S100A11 and MMP9 were detected in patients with EOC, patients with benign ovarian tumor, and healthy women. The correlation between the two markers and clinicopathological characteristics of ovarian cancer was analysed. Results The serum levels of S100A11 and MMP-9 in patients with EOC were higher than those in patients with benign ovarian tumor and in healthy women, and the expression levels of S100A11 and MMP-9 were positively correlated. S100A11 and MMP-9 were correlated with tumor staging, postoperative residual foci, ascites volume, serum CA125 level, chemotherapy response, and lymph node metastasis, while S100A11 and MMP-9 were not associated with the bilevel classification, histological type, age, and degree of differentiation. Conclusion S100A11 and MMP-9 were both highly expressed in the serum of patients with EOC and were associated with cancer development, invasion, and metastasis. Therefore, they can be used as an important reference maker in the diagnosis and treatment of ovarian cancer.
Collapse
|
24
|
Cecil D, Park KH, Curtis B, Corulli L, Disis MN. Type I T cells sensitize treatment refractory tumors to chemotherapy through inhibition of oncogenic signaling pathways. J Immunother Cancer 2021; 9:e002355. [PMID: 33762321 PMCID: PMC7993179 DOI: 10.1136/jitc-2021-002355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The most common clinical outcome observed after treatment with immune checkpoint inhibitor antibodies is disease stabilization. Using vaccines to generate high levels of tumor antigen-specific T-helper 1 (Th1), we show that tumors not eradicated by vaccination demonstrate prolonged disease stabilization. We evaluated the mechanism by which type I T cells inhibit disease progression and potentially influence the subsequent clinical response to standard therapy in treatment refractory cancers. METHODS We employed a meta-analysis of studies with tumor growth from four different vaccines in two different mammary cancer models. The T-cell subtype and cytokine essential for vaccine-induced tumor inhibition was determined by in vivo neutralization studies and immunohistochemistry. The role of interferon gamma (IFN-γ) in receptor tyrosine kinase and downstream signaling was determined by immunoblotting. The role of suppressor of cytokine signaling 1 (SOCS1) on IFN-γ signaling was evaluated on SOCS1-silenced cells with immunoblotting and immunoprecipitation. The effect of vaccination on growth factor receptor signaling pathways, performed in both luminal (TgMMTVneu) and basal (C3(1)-Tag) mammary cancer models treated with paclitaxel or an anti-HER2-neu monoclonal antibody were assessed via immunoblotting. RESULTS Immunization with an epitope-based vaccine targeting a representative tumor antigen resulted in elevated tumor trafficking Tbet+CD4 T cells, decreased tumor proliferation and increased apoptosis compared with control vaccinated mice. The resulting disease stabilization was dependent on IFN-γ-secreting CD4+ T cells. In the presence of excess IFN-γ, SOCS1 became upregulated in tumor cells, bound insulin receptor, insulin like growth factor receptor 1 and epidermal growth factor receptor resulting in profound oncogenic signaling inhibition. Silencing SOCS1 restored growth factor receptor signaling and proliferation and prevented cell death. Similar signaling perturbations were detected in vaccinated mice developing antigen-specific Th1 cells. Vaccination synergized with standard therapies and restored disease sensitivity to treatment with both a neu-specific antibody and paclitaxel in TgMMTVneu and to paclitaxel in C3(1)-Tag. Combination of vaccination and chemotherapy or biological therapy was more effective than monotherapy alone in either model and resulted in complete resolution of disease in some individuals. CONCLUSIONS These data suggest the clinical activity of type I T cells extends beyond direct tumor killing and immune therapies designed to increase type I T cells and could be integrated into standard chemotherapy regimens to enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Denise Cecil
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA, USA
| | | | - Benjamin Curtis
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA, USA
| | - Lauren Corulli
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA, USA
| | - Mary Nora Disis
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
25
|
Larionova I, Tuguzbaeva G, Ponomaryova A, Stakheyeva M, Cherdyntseva N, Pavlov V, Choinzonov E, Kzhyshkowska J. Tumor-Associated Macrophages in Human Breast, Colorectal, Lung, Ovarian and Prostate Cancers. Front Oncol 2020; 10:566511. [PMID: 33194645 PMCID: PMC7642726 DOI: 10.3389/fonc.2020.566511] [Citation(s) in RCA: 227] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are major innate immune cells that constitute up to 50% of the cell mass of human tumors. TAMs are highly heterogeneous cells that originate from resident tissue-specific macrophages and from newly recruited monocytes. TAMs' variability strongly depends on cancer type, stage, and intratumor heterogeneity. Majority of TAMs are programmed by tumor microenvironment to support primary tumor growth and metastatic spread. However, TAMs can also restrict tumor growth and metastasis. In this review, we summarized the knowledge about the role of TAMs in tumor growth, metastasis and in the response to cancer therapy in patients with five aggressive types of cancer: breast, colorectal, lung, ovarian, and prostate cancers that are frequently metastasize into distant organs resulting in high mortality of the patients. Two major TAM parameters are applied for the evaluation of TAM correlation with the cancer progression: total amount of TAMs and specific phenotype of TAMs identified by functional biomarkers. We summarized the data generated in the wide range of international patient cohorts on the correlation of TAMs with clinical and pathological parameters of tumor progression including lymphatic and hematogenous metastasis, recurrence, survival, therapy efficiency. We described currently available biomarkers for TAMs that can be measured in patients' samples (tumor tissue and blood). CD68 is the major biomarker for the quantification of total TAM amounts, while transmembrane receptors (stabilin-1, CD163, CD206, CD204, MARCO) and secreted chitinase-like proteins (YKL-39, YKL-40) are used as biomarkers for the functional TAM polarization. We also considered that specific role of TAMs in tumor progression can depend on the localization in the intratumoral compartments. We have made the conclusion for the role of TAMs in primary tumor growth, metastasis, and therapy sensitivity for breast, colorectal, lung, ovarian, and prostate cancers. In contrast to other cancer types, majority of clinical studies indicate that TAMs in colorectal cancer have protective role for the patient and interfere with primary tumor growth and metastasis. The accumulated data are essential for using TAMs as biomarkers and therapeutic targets to develop cancer-specific immunotherapy and to design efficient combinations of traditional therapy and new immunomodulatory approaches.
Collapse
Affiliation(s)
- Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Gulnara Tuguzbaeva
- Department of Pathophysiology, Bashkir State Medical University, Ufa, Russia
| | - Anastasia Ponomaryova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Marina Stakheyeva
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Nadezhda Cherdyntseva
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Valentin Pavlov
- Department of Urology, Bashkir State Medical University, Ufa, Russia
| | - Evgeniy Choinzonov
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Julia Kzhyshkowska
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg—Hessen, Mannheim, Germany
| |
Collapse
|
26
|
Martin-Lluesma S, Graciotti M, Grimm AJ, Boudousquié C, Chiang CL, Kandalaft LE. Are dendritic cells the most appropriate therapeutic vaccine for patients with ovarian cancer? Curr Opin Biotechnol 2020; 65:190-196. [DOI: 10.1016/j.copbio.2020.03.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/10/2020] [Accepted: 03/19/2020] [Indexed: 12/14/2022]
|
27
|
The influence of secreted factors and extracellular vesicles in ovarian cancer metastasis. EJC Suppl 2020; 15:38-48. [PMID: 33240441 PMCID: PMC7573474 DOI: 10.1016/j.ejcsup.2019.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 09/02/2019] [Accepted: 09/15/2019] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer cells mainly metastasise within the peritoneal cavity, the lethal consequence of tumour progression in this cancer type. Classically, changes in tumour cells, such as epithelial to mesenchymal transition, involve the down-regulatinon of E-cadherin, activation of extracellular proteases and integrin-mediated adhesion. However, our current understanding of ovarian tumour progression suggests the implication of both intrinsic and extrinsic factors. It has been proposed that ovarian cancer metastases are a consequence of the crosstalk between cancer cells and the tumour microenvironment by soluble factors and extracellular vesicles. Characterisation of the alterations in both the tumour cells and the surrounding microenvironment has emerged as a new research field to understand ovarian cancer metastasis. In this mini review, we will summarise the most recent findings, focusing our attention on the role of secreted factors and extracellular vesicles in ovarian cancer metastasis. During ovarian cancer metastasis, tumour cells metastasise in the mesothelium as primarily ‘soil’ for ovarian cancer ‘seeds’. Soluble factors and extracellular vesicles secreted by tumor cells are involved in the generation of the pre-metastatic niche. Cancer-associated fibroblasts (CAFs) represent the majority of stromal cells in various types of human carcinoma, including ovarian cancer. Analysis of early metastasis to the omentum indicates that ovarian cancer cells rely on the interaction with immune cells such as macrophages. Liquid biopsy analyses in ovarian cancer may help to define novel biomarkers improving patient survival and reduce lethality.
Collapse
|
28
|
Adams SF, Grimm AJ, Chiang CLL, Mookerjee A, Flies D, Jean S, McCann GA, Michaux J, Pak H, Huber F, Neal C, Dangaj D, Bassani-Sternberg M, Rusakiewicz S, Facciabene A, Coukos G, Gimotty PA, Kandalaft LE. Rapid tumor vaccine using Toll-like receptor-activated ovarian cancer ascites monocytes. J Immunother Cancer 2020; 8:jitc-2020-000875. [PMID: 32817208 PMCID: PMC7430560 DOI: 10.1136/jitc-2020-000875] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Background Novel therapeutic strategies in ovarian cancer (OC) are needed as the survival rate remains dismally low. Although dendritic cell-based cancer vaccines are effective in eliciting therapeutic responses, their complex and costly manufacturing process hampers their full clinical utility outside specialized clinics. Here, we describe a novel approach of generating a rapid and effective cancer vaccine using ascites-derived monocytes for treating OC. Methods Using the ID8 mouse ovarian tumor model and OC patient samples, we isolated ascites monocytes and evaluated them with flow cytometry, Luminex cytokine and chemokine array analysis, ex vivo cocultures with T cells, in vivo tumor challenge and T cell transfer experiments, RNA-sequencing and mass spectrometry. Results We demonstrated the feasibility of isolating ascites monocytes and restoring their ability to function as bona fide antigen-presenting cells (APCs) with Toll-like receptor (TLR) 4 lipopolysaccharide and TLR9 CpG-oligonucleotides, and a blocking antibody to interleukin-10 receptor (IL-10R Ab) in the ID8 model. The ascites monocytes were laden with tumor antigens at a steady state in vivo. After a short 48 hours activation, they upregulated maturation markers (CD80, CD86 and MHC class I) and demonstrated strong ex vivo T cell stimulatory potential and effectively suppressed tumor and malignant ascites in vivo. They also induced protective long-term T cell memory responses. To evaluate the translational potential of this approach, we isolated ascites monocytes from stage III/IV chemotherapy-naïve OC patients. Similarly, the human ascites monocytes presented tumor-associated antigens (TAAs), including MUC1, ERBB2, mesothelin, MAGE, PRAME, GPC3, PMEL and TP53 at a steady state. After a 48-hour treatment with TLR4 and IL-10R Ab, they efficiently stimulated oligoclonal tumor-associated lymphocytes (TALs) with strong reactivity against TAAs. Importantly, the activated ascites monocytes retained their ability to activate TALs in the presence of ascitic fluid. Conclusions Ascites monocytes are naturally loaded with tumor antigen and can perform as potent APCs following short ex vivo activation. This novel ascites APC vaccine can be rapidly prepared in 48 hours with a straightforward and affordable manufacturing process, and would be an attractive therapeutic vaccine for OC.
Collapse
Affiliation(s)
- Sarah F Adams
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Gynecologic Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| | - Alizée J Grimm
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Cheryl L-L Chiang
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Ananda Mookerjee
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dallas Flies
- Division of Gynecologic Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| | - Stephanie Jean
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Georgia A McCann
- Division of Gynecologic Oncology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Justine Michaux
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - HuiSong Pak
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Florian Huber
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Christopher Neal
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Denarda Dangaj
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Sylvie Rusakiewicz
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Andrea Facciabene
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - George Coukos
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Phyllis A Gimotty
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lana E Kandalaft
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland .,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| |
Collapse
|
29
|
Feng Y, Xiao M, Zhang Z, Cui R, Jiang X, Wang S, Bai H, Liu C, Zhang Z. Potential interaction between lysophosphatidic acid and tumor-associated macrophages in ovarian carcinoma. JOURNAL OF INFLAMMATION-LONDON 2020; 17:23. [PMID: 32774171 PMCID: PMC7405460 DOI: 10.1186/s12950-020-00254-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/30/2020] [Indexed: 02/06/2023]
Abstract
Ovarian carcinoma is the deadliest type of gynecological cancer. The unique tumor microenvironment enables specific and efficient metastasis, weakens immunological monitoring, and mediates drug resistance. Tumor associated macrophages (TAMs) are a crucial part of the TME and are involved in various aspects of tumor behavior. Lysophosphatidic acid (LPA) is elevated in the blood of ovarian carcinoma patients, as well as in the tumor tissues and ascites, which make it a useful biomarker and a potential therapeutic target. Recent studies have shown that LPA transforms monocytes into macrophages and regulates the formation of macrophages through the AKT/mTOR pathway, and PPAR γ is a major regulator of LPA-derived macrophages. In addition, TAMs synthesize and secrete LPA and express LPA receptor (LPAR) on the surface. With these data in mind, we hypothesize that LPA can convert monocytes directly into TAMs in the microenvironment of ovarian cancer. LPA may mediate TAM formation by activating the PI3K/AKT/mTOR signaling pathway through LPAR on the cell surface, which may also affect the function of PPAR γ, leading to increased LPA production by TAMs. Thus, LPA and TAMs form a vicious circle that affects the malignant behavior of ovarian cancer.
Collapse
Affiliation(s)
- Ying Feng
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Meizhu Xiao
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Zihan Zhang
- Department of Gynecology and Obstetrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ran Cui
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Xuan Jiang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Shuzhen Wang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Huimin Bai
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Chongdong Liu
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Zhenyu Zhang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| |
Collapse
|
30
|
Martinez A, Delord JP, Ayyoub M, Devaud C. Preclinical and Clinical Immunotherapeutic Strategies in Epithelial Ovarian Cancer. Cancers (Basel) 2020; 12:E1761. [PMID: 32630708 PMCID: PMC7409311 DOI: 10.3390/cancers12071761] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 12/25/2022] Open
Abstract
In the past 20 years, the immune system has increasingly been recognized as a major player in tumor cell control, leading to considerable advances in cancer treatment. While promising with regards to melanoma, renal cancer and non-small cell lung cancer, immunotherapy provides, for the time being, limited success in other cancers, including ovarian cancer, potentially due to insufficient immunogenicity or to a particularly immunosuppressive microenvironment. In this review, we provide a global description of the immune context of ovarian cancer, in particular epithelial ovarian cancer (EOC). We describe the adaptive and innate components involved in the EOC immune response, including infiltrating tumor-specific T lymphocytes, B lymphocytes, and natural killer and myeloid cells. In addition, we highlight the rationale behind the use of EOC preclinical mouse models to assess resistance to immunotherapy, and we summarize the main preclinical studies that yielded anti-EOC immunotherapeutic strategies. Finally, we focus on major published or ongoing immunotherapy clinical trials concerning EOC.
Collapse
Affiliation(s)
- Alejandra Martinez
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé Et de la Recherche Médicale (INSERM) Unité 1037, 31037 Toulouse, France; (A.M.); (J.-P.D.); (M.A.)
- Department of Surgery, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse (IUCT), 31037 Toulouse, France
| | - Jean-Pierre Delord
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé Et de la Recherche Médicale (INSERM) Unité 1037, 31037 Toulouse, France; (A.M.); (J.-P.D.); (M.A.)
- Department of Medical Oncology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 31037 Toulouse, France
- Université Toulouse III Paul Sabatier, 31037 Toulouse, France
| | - Maha Ayyoub
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé Et de la Recherche Médicale (INSERM) Unité 1037, 31037 Toulouse, France; (A.M.); (J.-P.D.); (M.A.)
- Université Toulouse III Paul Sabatier, 31037 Toulouse, France
- Immune Monitoring Core Facility, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 31037 Toulouse, France
| | - Christel Devaud
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé Et de la Recherche Médicale (INSERM) Unité 1037, 31037 Toulouse, France; (A.M.); (J.-P.D.); (M.A.)
- Immune Monitoring Core Facility, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 31037 Toulouse, France
| |
Collapse
|
31
|
The Role of Tumor-Associated Macrophages in the Progression and Chemoresistance of Ovarian Cancer. Cells 2020; 9:cells9051299. [PMID: 32456078 PMCID: PMC7290435 DOI: 10.3390/cells9051299] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 01/11/2023] Open
Abstract
Tumor-associated macrophages (TAMs) constitute the main population of immune cells present in the ovarian tumor microenvironment. These cells are characterized by high plasticity and can be easily polarized by colony-stimulating factor-1, which is released by tumor cells, into an immunosuppressive M2-like phenotype. These cells are strongly implicated in both the progression and chemoresistance of ovarian cancer. The main pro-tumoral function of M2-like TAMs is the secretion of a variety of cytokines, chemokines, enzymes and exosomes that reach microRNAs, directly inducing the invasion potential and chemoresistance of ovarian cancer cells by triggering their pro-survival signaling pathways. The M2-like TAMs are also important players in the metastasis of ovarian cancer cells in the peritoneum through their assistance in spheroid formation and attachment of cancer cells to the metastatic area—the omentum. Moreover, TAMs interplay with other immune cells, such as lymphocytes, natural killer cells, and dendritic cells, to inhibit their responsiveness, resulting in the development of immunosuppression. The detrimental character of the M2-like type of TAMs in ovarian tumors has been confirmed by a number of studies, demonstrating the positive correlation between their high level in tumors and low overall survival of patients.
Collapse
|
32
|
Steitz AM, Steffes A, Finkernagel F, Unger A, Sommerfeld L, Jansen JM, Wagner U, Graumann J, Müller R, Reinartz S. Tumor-associated macrophages promote ovarian cancer cell migration by secreting transforming growth factor beta induced (TGFBI) and tenascin C. Cell Death Dis 2020; 11:249. [PMID: 32312959 PMCID: PMC7171168 DOI: 10.1038/s41419-020-2438-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 12/13/2022]
Abstract
A central and unique aspect of high-grade serous ovarian carcinoma (HGSC) is the extensive transcoelomic spreading of tumor cell via the peritoneal fluid or malignant ascites. We and others identified tumor-associated macrophages (TAM) in the ascites as promoters of metastasis-associated processes like extracellular matrix (ECM) remodeling, tumor cell migration, adhesion, and invasion. The precise mechanisms and mediators involved in these functions of TAM are, however, largely unknown. We observed that HGSC migration is promoted by soluble mediators from ascites-derived TAM, which can be emulated by conditioned medium from monocyte-derived macrophages (MDM) differentiated in ascites to TAM-like asc-MDM. A similar effect was observed with IL-10-induced alternatively activated m2c-MDM but not with LPS/IFNγ-induced inflammatory m1-MDM. These observations provided the basis for deconvolution of the complex TAM secretome by performing comparative secretome analysis of matched triplets of different MDM phenotypes with different pro-migratory properties (asc-MDM, m2c-MDM, m1-MDM). Mass spectrometric analysis identified an overlapping set of nine proteins secreted by both asc-MDM and m2c-MDM, but not by m1-MDM. Of these, three proteins, i.e., transforming growth factor beta-induced (TGFBI) protein, tenascin C (TNC), and fibronectin (FN1), have been associated with migration-related functions. Intriguingly, increased ascites concentrations of TGFBI, TNC, and fibronectin were associated with short progression-free survival. Furthermore, transcriptome and secretome analyses point to TAM as major producers of these proteins, further supporting an essential role for TAM in promoting HGSC progression. Consistent with this hypothesis, we were able to demonstrate that the migration-inducing potential of asc-MDM and m2c-MDM secretomes is inhibited, at least partially, by neutralizing antibodies against TGFBI and TNC or siRNA-mediated silencing of TGFBI expression. In conclusion, the present study provides the first experimental evidence that TAM-derived TGFBI and TNC in ascites promote HGSC progression.
Collapse
Affiliation(s)
- Anna Mary Steitz
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Alina Steffes
- Clinic for Gynecology, Gynecologic Oncology and Endocrinology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Florian Finkernagel
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Annika Unger
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Leah Sommerfeld
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Julia M Jansen
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital Giessen and Marburg (UKGM), Marburg, Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital Giessen and Marburg (UKGM), Marburg, Germany
| | - Johannes Graumann
- Biomolecular Mass Spectrometry, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany.,The German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany.
| | - Silke Reinartz
- Clinic for Gynecology, Gynecologic Oncology and Endocrinology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| |
Collapse
|
33
|
Tong DL, Kempsell KE, Szakmany T, Ball G. Development of a Bioinformatics Framework for Identification and Validation of Genomic Biomarkers and Key Immunopathology Processes and Controllers in Infectious and Non-infectious Severe Inflammatory Response Syndrome. Front Immunol 2020; 11:380. [PMID: 32318053 PMCID: PMC7147506 DOI: 10.3389/fimmu.2020.00380] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 02/17/2020] [Indexed: 12/12/2022] Open
Abstract
Sepsis is defined as dysregulated host response caused by systemic infection, leading to organ failure. It is a life-threatening condition, often requiring admission to an intensive care unit (ICU). The causative agents and processes involved are multifactorial but are characterized by an overarching inflammatory response, sharing elements in common with severe inflammatory response syndrome (SIRS) of non-infectious origin. Sepsis presents with a range of pathophysiological and genetic features which make clinical differentiation from SIRS very challenging. This may reflect a poor understanding of the key gene inter-activities and/or pathway associations underlying these disease processes. Improved understanding is critical for early differential recognition of sepsis and SIRS and to improve patient management and clinical outcomes. Judicious selection of gene biomarkers suitable for development of diagnostic tests/testing could make differentiation of sepsis and SIRS feasible. Here we describe a methodologic framework for the identification and validation of biomarkers in SIRS, sepsis and septic shock patients, using a 2-tier gene screening, artificial neural network (ANN) data mining technique, using previously published gene expression datasets. Eight key hub markers have been identified which may delineate distinct, core disease processes and which show potential for informing underlying immunological and pathological processes and thus patient stratification and treatment. These do not show sufficient fold change differences between the different disease states to be useful as primary diagnostic biomarkers, but are instrumental in identifying candidate pathways and other associated biomarkers for further exploration.
Collapse
Affiliation(s)
- Dong Ling Tong
- Artificial Intelligence Laboratory, Faculty of Engineering and Computing, First City University College, Petaling Jaya, Malaysia.,School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Karen E Kempsell
- Public Health England, National Infection Service, Porton Down, Salisbury, United Kingdom
| | - Tamas Szakmany
- Department of Anaesthesia Intensive Care and Pain Medicine, Division of Population Medicine, Cardiff University, Cardiff, United Kingdom
| | - Graham Ball
- School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
34
|
Yin M, Shen J, Yu S, Fei J, Zhu X, Zhao J, Zhai L, Sadhukhan A, Zhou J. Tumor-Associated Macrophages (TAMs): A Critical Activator In Ovarian Cancer Metastasis. Onco Targets Ther 2019; 12:8687-8699. [PMID: 31695427 PMCID: PMC6814357 DOI: 10.2147/ott.s216355] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/24/2019] [Indexed: 12/13/2022] Open
Abstract
Tumor-associated macrophages (TAMs) that appear in every stage of cancer progression are usually tumor-promoting cells and are present abundantly in the tumor-associated microenvironment. In ovarian cancer, the overall and intratumoral M1/M2 ratio is a relatively efficient TAM parameter for predicting the prognosis of patients, especially for serous tissue type cancer. TAMs exhibit immunological checkpoint modulators, such as the B7 family and programmed death-ligand 1 (PD-L1), and play a key role in the development, metastasis and invasion of ovarian cancer, but the underlying mechanism is barely understood. Ovarian cancer is a severe gynecological malignancy with high mortality. Ovarian cancer-associated death can primarily be attributed to cancer metastasis. The majority of patients are diagnosed with wide dissemination in the peritoneum and omentum, limiting the effectiveness of surgery and chemotherapy. In addition, unlike other well-documented cancers, metastasis through vasculature is not a usual dissemination pathway in ovarian cancer. This review sheds light on TAMs and the main process and mechanism of ovarian cancer metastasis.
Collapse
Affiliation(s)
- Meichen Yin
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Jiayu Shen
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Shuqian Yu
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Jing Fei
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Xiaoqing Zhu
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Jiayao Zhao
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Lingyun Zhai
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Annapurna Sadhukhan
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
35
|
Hu BL, Xie MZ, Li KZ, Li JL, Gui YC, Xu JW. Genome-wide analysis to identify a novel distant metastasis-related gene signature predicting survival in patients with gastric cancer. Biomed Pharmacother 2019; 117:109159. [DOI: 10.1016/j.biopha.2019.109159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 12/29/2022] Open
|
36
|
Yin S, Du J, Zhang J, Zhang X, Ma K. Identification of Key Genes and Pathway for Ovarian Neoplasms Using the OVDM1 Cell Line Based on Bioinformatics Analysis. Med Sci Monit 2019; 25:4305-4313. [PMID: 31177266 PMCID: PMC6582691 DOI: 10.12659/msm.915422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Ovarian neoplasms are the fifth most common cancer affecting the health of women, and they are the most lethal gynecologic malignancies; however, the etiology of ovarian neoplasms remains largely unknown. There is an urgent need to further broaden the understanding of the development mechanism of ovarian neoplasms through in vitro research using different cell lines. Material/Methods To screen the differentially expressed genes (DEGs) that may play critical roles in OVDM1 (an ovarian cancer cell line), the public microarray data (GSE70264) were downloaded and screened for DEGs. Then, Gene Ontology (GO) function analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed. To screen hub genes, the protein–protein interaction network was constructed. The expression level and survival analysis of hub genes in patients with ovarian neoplasms were also analyzed. Results There were 79 upregulated and 926 downregulated DEGs detected, and the biological processes of the GO analysis were enriched in extracellular matrix organization, extracellular structure organization, and chromosome segregation, whereas, the KEGG pathway analysis was enriched in cell cycle and cell adhesion molecules. The hub gene BIRC5, which might play a key role in ovarian neoplasms, was further screened. Conclusions The present study could deepen the understanding of the molecular mechanism of ovarian neoplasms using the OVDM1 cell line, which could be useful in developing clinical treatments of ovarian neoplasms.
Collapse
Affiliation(s)
- Songna Yin
- Medical School, Yan'an University, Yan'an, Shaanxi, China (mainland)
| | - Juan Du
- Medical School, Yan'an University, Yan'an, Shaanxi, China (mainland)
| | - Jie Zhang
- Medical School, Yan'an University, Yan'an, Shaanxi, China (mainland)
| | - Xiang Zhang
- Medical School, Yan'an University, Yan'an, Shaanxi, China (mainland)
| | - Ke Ma
- Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| |
Collapse
|
37
|
Aquila I, Ricci P, Oliverio A, Gratteri S. Role of the body mass index in the genesis of ascites in ovarian cancer: a forensic case and review of the literature. BMJ Case Rep 2018; 11:11/1/e226491. [PMID: 30567893 DOI: 10.1136/bcr-2018-226491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The ovarian tumour is the seventh female cancer for incidence. In the advanced stages of cancer, tumour cells nourish on the peritoneal serous causing carcinomatosis and peritoneal function abnormalities with liquid build-up inside it. Ascites from peritoneal carcinomatosis is common in patients with ovarian cancer. An obese woman suffering from ovarian cancer was found dead in her home from secondary cardio-respiratory arrest due to Multiple Organ Failure (MOF). An autopsy was performed. The abdominal incision showed an ascitic fluid outflow about 20 litres in volume and a flood about one metre and half. An association between obesity and intraperitoneal fluid volume secondary to peritoneal carcinomatosis has been demonstrated. This finding could improve the prognosis of patients through actions aimed to reduce body weight.
Collapse
Affiliation(s)
- Isabella Aquila
- Institute of Legale Medicine, University Magna Graecia of Catanzaro, Catanzaro, Catanzaro, Italy
| | - Pietrantonio Ricci
- Institute of Legale Medicine, University Magna Graecia of Catanzaro, Catanzaro, Catanzaro, Italy
| | - Alessandra Oliverio
- Institute of Legale Medicine, University Magna Graecia of Catanzaro, Catanzaro, Catanzaro, Italy
| | - Santo Gratteri
- Institute of Legal Medicine, AO MaterDomini Catanzaro, Catanzaro, Italy
| |
Collapse
|
38
|
Reinartz S, Lieber S, Pesek J, Brandt DT, Asafova A, Finkernagel F, Watzer B, Nockher WA, Nist A, Stiewe T, Jansen JM, Wagner U, Konzer A, Graumann J, Grosse R, Worzfeld T, Müller-Brüsselbach S, Müller R. Cell type-selective pathways and clinical associations of lysophosphatidic acid biosynthesis and signaling in the ovarian cancer microenvironment. Mol Oncol 2018; 13:185-201. [PMID: 30353652 PMCID: PMC6360368 DOI: 10.1002/1878-0261.12396] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 10/02/2018] [Accepted: 10/14/2018] [Indexed: 12/18/2022] Open
Abstract
The peritoneal fluid of ovarian carcinoma patients promotes cancer cell invasion and metastatic spread with lysophosphatidic acid (LPA) as a potentially crucial mediator. However, the origin of LPA in ascites and the clinical relevance of individual LPA species have not been addressed. Here, we show that the levels of multiple acyl‐LPA species are strongly elevated in ascites versus plasma and are associated with short relapse‐free survival. Data derived from transcriptome and secretome analyses of primary ascite‐derived cells indicate that (a) the major route of LPA synthesis is the consecutive action of a secretory phospholipase A2 (PLA2) and autotaxin, (b) that the components of this pathway are coordinately upregulated in ascites, and (c) that CD163+CD206+ tumor‐associated macrophages play an essential role as main producers of PLA2G7 and autotaxin. The latter conclusion is consistent with mass spectrometry‐based metabolomic analyses of conditioned medium from ascites cells, which showed that tumor‐associated macrophages, but not tumor cells, are able to produce 20:4 acyl‐LPA in lipid‐free medium. Furthermore, our transcriptomic data revealed that LPA receptor (LPAR) genes are expressed in a clearly cell type‐selective manner: While tumor cells express predominantly LPAR1‐3, macrophages and T cells also express LPAR5 and LPAR6 at high levels, pointing to cell type‐selective LPA signaling pathways. RNA profiling identified cytokines linked to cell motility and migration as the most conspicuous class of LPA‐induced genes in macrophages, suggesting that LPA exerts protumorigenic properties at least in part via the tumor secretome.
Collapse
Affiliation(s)
- Silke Reinartz
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Marburg, Germany
| | - Sonja Lieber
- Center for Tumor Biology and Immunology (ZTI), Institute of Molecular Biology and Tumor Research (IMT), Marburg, Germany
| | - Jelena Pesek
- Metabolomics Core Facility, Philipps University, Marburg, Germany
| | | | - Alina Asafova
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Marburg, Germany.,Center for Tumor Biology and Immunology (ZTI), Institute of Molecular Biology and Tumor Research (IMT), Marburg, Germany
| | - Florian Finkernagel
- Center for Tumor Biology and Immunology (ZTI), Institute of Molecular Biology and Tumor Research (IMT), Marburg, Germany
| | - Bernard Watzer
- Metabolomics Core Facility, Philipps University, Marburg, Germany
| | | | - Andrea Nist
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Julia M Jansen
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, UKGM, Marburg, Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, UKGM, Marburg, Germany
| | - Anne Konzer
- Biomolecular Mass Spectrometry, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Kerckhoff Klinik, Bad Nauheim, Germany
| | - Johannes Graumann
- Biomolecular Mass Spectrometry, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Kerckhoff Klinik, Bad Nauheim, Germany
| | | | - Thomas Worzfeld
- Institute of Pharmacology, Marburg, Germany.,Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Sabine Müller-Brüsselbach
- Center for Tumor Biology and Immunology (ZTI), Institute of Molecular Biology and Tumor Research (IMT), Marburg, Germany
| | - Rolf Müller
- Center for Tumor Biology and Immunology (ZTI), Institute of Molecular Biology and Tumor Research (IMT), Marburg, Germany
| |
Collapse
|
39
|
Curigliano G. Gyneco-oncological genomics and emerging biomarkers for cancer treatment with immune-checkpoint inhibitors. Semin Cancer Biol 2018; 52:253-258. [DOI: 10.1016/j.semcancer.2018.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 05/14/2018] [Indexed: 12/20/2022]
|
40
|
Gupta V, Yull F, Khabele D. Bipolar Tumor-Associated Macrophages in Ovarian Cancer as Targets for Therapy. Cancers (Basel) 2018. [PMID: 30274280 DOI: 10.3390/cancers10100366] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ovarian cancer, a rare but fatal disease, has been a challenging area in the field of gynecological cancer. Ovarian cancer is characterized by peritoneal metastasis, which is facilitated by a cross-talk between tumor cells and other cells in the tumor microenvironment (TME). In epithelial ovarian cancer, tumor-associated macrophages (TAMs) constitute over 50% of cells in the peritoneal TME and malignant ascites, and are potential targets for therapy. Here, we review the bipolar nature of TAMs and the evolving strategies to target TAMs in ovarian cancer.
Collapse
Affiliation(s)
- Vijayalaxmi Gupta
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Fiona Yull
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.
- Vanderbilt-Ingram Cancer Center, Nashville, TN 37232, USA.
| | - Dineo Khabele
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
- The University of Kansas Cancer Center, Kansas City, KS 66160, USA.
| |
Collapse
|
41
|
Bipolar Tumor-Associated Macrophages in Ovarian Cancer as Targets for Therapy. Cancers (Basel) 2018; 10:cancers10100366. [PMID: 30274280 PMCID: PMC6210537 DOI: 10.3390/cancers10100366] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/22/2018] [Accepted: 09/23/2018] [Indexed: 12/31/2022] Open
Abstract
Ovarian cancer, a rare but fatal disease, has been a challenging area in the field of gynecological cancer. Ovarian cancer is characterized by peritoneal metastasis, which is facilitated by a cross-talk between tumor cells and other cells in the tumor microenvironment (TME). In epithelial ovarian cancer, tumor-associated macrophages (TAMs) constitute over 50% of cells in the peritoneal TME and malignant ascites, and are potential targets for therapy. Here, we review the bipolar nature of TAMs and the evolving strategies to target TAMs in ovarian cancer.
Collapse
|
42
|
Gupta V, Yull F, Khabele D. Bipolar Tumor-Associated Macrophages in Ovarian Cancer as Targets for Therapy. Cancers (Basel) 2018. [PMID: 30274280 DOI: 10.3390/cancers10100366]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Ovarian cancer, a rare but fatal disease, has been a challenging area in the field of gynecological cancer. Ovarian cancer is characterized by peritoneal metastasis, which is facilitated by a cross-talk between tumor cells and other cells in the tumor microenvironment (TME). In epithelial ovarian cancer, tumor-associated macrophages (TAMs) constitute over 50% of cells in the peritoneal TME and malignant ascites, and are potential targets for therapy. Here, we review the bipolar nature of TAMs and the evolving strategies to target TAMs in ovarian cancer.
Collapse
Affiliation(s)
- Vijayalaxmi Gupta
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Fiona Yull
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.
- Vanderbilt-Ingram Cancer Center, Nashville, TN 37232, USA.
| | - Dineo Khabele
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
- The University of Kansas Cancer Center, Kansas City, KS 66160, USA.
| |
Collapse
|
43
|
Unger A, Finkernagel F, Hoffmann N, Neuhaus F, Joos B, Nist A, Stiewe T, Visekruna A, Wagner U, Reinartz S, Müller-Brüsselbach S, Müller R, Adhikary T. Chromatin Binding of c-REL and p65 Is Not Limiting for Macrophage IL12B Transcription During Immediate Suppression by Ovarian Carcinoma Ascites. Front Immunol 2018; 9:1425. [PMID: 29997615 PMCID: PMC6030372 DOI: 10.3389/fimmu.2018.01425] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 06/08/2018] [Indexed: 12/14/2022] Open
Abstract
Tumors frequently exploit homeostatic mechanisms that suppress expression of IL-12, a central mediator of inflammatory and anti-tumor responses. The p40 subunit of the IL-12 heterodimer, encoded by IL12B, is limiting for these functions. Ovarian carcinoma patients frequently produce ascites which exerts immunosuppression by means of soluble factors. The NFκB pathway is necessary for transcription of IL12B, which is not expressed in macrophages freshly isolated from ascites. This raises the possibility that ascites prevents IL12B expression by perturbing NFκB binding to chromatin. Here, we show that ascites-mediated suppression of IL12B induction by LPS plus IFNγ in primary human macrophages is rapid, and that suppression can be reversible after ascites withdrawal. Nuclear translocation of the NFκB transcription factors c-REL and p65 was strongly reduced by ascites. Surprisingly, however, their binding to the IL12B locus and to CXCL10, a second NFκB target gene, was unaltered, and the induction of CXCL10 transcription was not suppressed by ascites. These findings indicate that, despite its reduced nuclear translocation, NFκB function is not generally impaired by ascites, suggesting that ascites-borne signals target additional pathways to suppress IL12B induction. Consistent with these data, IL-10, a clinically relevant constituent of ascites and negative regulator of NFκB translocation, only partially recapitulated IL12B suppression by ascites. Finally, restoration of a defective IL-12 response by appropriate culture conditions was observed only in macrophages from a subset of donors, which may have important implications for the understanding of patient-specific immune responses.
Collapse
Affiliation(s)
- Annika Unger
- Institute for Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| | - Florian Finkernagel
- Institute for Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| | - Nathalie Hoffmann
- Experimental Tumor Research Group, Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| | - Felix Neuhaus
- Institute for Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| | - Barbara Joos
- Institute for Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, ZTI, Philipps University of Marburg, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, ZTI, Philipps University of Marburg, Marburg, Germany
| | - Alexander Visekruna
- Institute for Medical Microbiology and Hygiene, Biomedical Research Center (BMFZ), Philipps University of Marburg, Marburg, Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Philipps University of Marburg, Marburg, Germany
| | - Silke Reinartz
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, ZTI, Philipps University of Marburg, Marburg, Germany
| | - Sabine Müller-Brüsselbach
- Institute for Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| | - Rolf Müller
- Institute for Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| | - Till Adhikary
- Institute for Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| |
Collapse
|
44
|
Tang M, Liu B, Bu X, Zhao P. Cross-talk between ovarian cancer cells and macrophages through periostin promotes macrophage recruitment. Cancer Sci 2018. [PMID: 29527764 PMCID: PMC5980394 DOI: 10.1111/cas.13567] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Tumor‐associated macrophages (TAMs) contribute to tumor progression, but it is not clear how they are recruited to tumor sites. Here we showed that periostin (POSTN) was present at high levels in ovarian cancer ascetic fluids and was correlated with CD163+TAMs. The high POSTN level and macrophage infiltration were inversely associated with relapse‐free survival for ovarian cancer patients. In vitro studies showed that coculture with macrophages significantly increased POSTN production in ovarian cancer cells. Further investigation found that POSTN production in ovarian cancer cells was promoted by transforming growth factor‐β generated by macrophages. Moreover, siRNA of POSTN and POSTN neutralizing antibody treatment showed that ovarian cancer cell‐derived POSTN promoted the recruitment of macrophages and modulated their cytokine secretion profile. Collectively, these data indicated that POSTN was an important factor for macrophage recruitment in the tumor microenvironment and is involved in the interactions between macrophages and ovarian cancer cells.
Collapse
Affiliation(s)
- Meng Tang
- Department of Thoracic Surgery, Qingdao Central Hospital, The Second Clinical Hospital of Qingdao University, Qingdao, China
| | - Bingji Liu
- Department of Nuclear Medicine, Qingdao Central Hospital, The Second Clinical Hospital of Qingdao University, Qingdao, China
| | - Xiaocui Bu
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China
| | - Peng Zhao
- Biotherapy Center, Qingdao Central Hospital, The Second Clinical Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
45
|
Worzfeld T, Finkernagel F, Reinartz S, Konzer A, Adhikary T, Nist A, Stiewe T, Wagner U, Looso M, Graumann J, Müller R. Proteotranscriptomics Reveal Signaling Networks in the Ovarian Cancer Microenvironment. Mol Cell Proteomics 2017; 17:270-289. [PMID: 29141914 PMCID: PMC5795391 DOI: 10.1074/mcp.ra117.000400] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Indexed: 01/17/2023] Open
Abstract
Ovarian cancer is characterized by early transcoelomic metastatic spread via the peritoneal fluid, where tumor cell spheroids (TU), tumor-associated T cells (TAT), and macrophages (TAM) create a unique microenvironment promoting cancer progression, chemoresistance, and immunosuppression. However, the underlying signaling mechanisms remain largely obscure. To chart these signaling networks, we performed comprehensive proteomic and transcriptomic analyses of TU, TAT, and TAM from ascites of ovarian cancer patients. We identify multiple intercellular signaling pathways driven by protein or lipid mediators that are associated with clinical outcome. Beyond cytokines, chemokines and growth factors, these include proteins of the extracellular matrix, immune checkpoint regulators, complement factors, and a prominent network of axon guidance molecules of the ephrin, semaphorin, and slit families. Intriguingly, both TU and TAM from patients with a predicted short survival selectively produce mediators supporting prometastatic events, including matrix remodeling, stemness, invasion, angiogenesis, and immunosuppression, whereas TAM associated with a longer survival express cytokines linked to effector T-cell chemoattraction and activation. In summary, our study uncovers previously unrecognized signaling networks in the ovarian cancer microenvironment that are of potential clinical relevance.
Collapse
Affiliation(s)
- Thomas Worzfeld
- From the ‡Institute of Pharmacology, Biochemical-Pharmacological Center (BPC), Philipps University, Marburg, Germany 35043; .,§Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany 61231
| | - Florian Finkernagel
- ¶Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany 35043
| | - Silke Reinartz
- ‖Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany 35043
| | - Anne Konzer
- **Biomolecular Mass Spectrometry, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany 61231
| | - Till Adhikary
- ¶Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany 35043
| | - Andrea Nist
- ‡‡Genomics Core Facility, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany 35043
| | - Thorsten Stiewe
- ‡‡Genomics Core Facility, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany 35043
| | - Uwe Wagner
- §§Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital of Giessen and Marburg (UKGM), Marburg, Germany 35043
| | - Mario Looso
- ¶¶Bioinformatics, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany 61231
| | - Johannes Graumann
- **Biomolecular Mass Spectrometry, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany 61231
| | - Rolf Müller
- ¶Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany 35043;
| |
Collapse
|