1
|
Zhang X, Xiong B, Cheng Y, Huang J, Xue J, Li X, Lu W, Zhu J, Wang L, Yang W, Cheng Z. Berberine inhibits metastasis of ovarian cancer by blocking lipid metabolism, alleviating aging of adipose tissue and increasing tumor infiltrating immune cells. Transl Oncol 2025; 56:102380. [PMID: 40252400 PMCID: PMC12033994 DOI: 10.1016/j.tranon.2025.102380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/20/2025] [Accepted: 03/24/2025] [Indexed: 04/21/2025] Open
Abstract
Extensive peritoneal metastasis and malignant ascites continue to pose substantial challenges in achieving favorable treatment outcomes for ovarian cancer. Berberine (BBR), an active component of numerous traditional Chinese herbs, has demonstrated potent anti - tumor effects across various malignancies, including ovarian cancer. In this study, we comprehensively evaluated the impact of BBR on the growth and metastasis of ovarian cancer both in vitro and in vivo. RNA - sequencing was employed to elucidate the underlying mechanisms. Specifically, we investigated lipid metabolism and mitochondrial function in ovarian cancer cells and mice, comparing BBR - treated and untreated groups. Additionally, CIBERSORT analysis and immunohistochemical (IHC) staining were utilized to confirm BBR's ability to enhance the infiltration of tumor-infiltrating immune cells into adipose tissue and improve the inflammatory tumor microenvironment. Our findings indicate that BBR significantly inhibits the growth and metastasis of ovarian cancer in vitro and in vivo. The effects can be attributed to two key processes. Firstly, BBR suppresses the lipid metabolism by downregulating lipid uptake related receptor CD36, lipid metabolic enzyme and mitochondrial function. Secondly, BBR alleviates the aging of adipose tissue and adipose derived stem cells (ADSCs), thereby decreasing the secretion of senescence-associated secretory phenotype (SASP). These ultimately lead to the increasing the improvement of tumor infiltrating immune cells, such as CD4⁺ helper T cells (CD3⁺CD4⁺) and cytotoxic T lymphocytes (CD3⁺CD8⁺), and inflammation in ovarian cancer tissue. Collectively, these findings suggested a potential therapeutic effect of BBR in the treatment of advanced ovarian cancer, particularly cases complicated by peritoneal metastasis and malignant ascites.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China; Department of Gynecology, Jing'an District Hospital of Traditional Chinese Medicine, Shanghai, 200072, PR China; Continuous Education College, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, PR China
| | - Bing Xiong
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China
| | - Yujie Cheng
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China
| | - Jimei Huang
- Continuous Education College, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, PR China
| | - Jiaying Xue
- Department of Gynecology, Jing'an District Hospital of Traditional Chinese Medicine, Shanghai, 200072, PR China; Continuous Education College, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, PR China
| | - Xiao Li
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China
| | - Wei Lu
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China
| | - Jihui Zhu
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China
| | - Lian Wang
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China.
| | - Weihong Yang
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China.
| | - Zhongping Cheng
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, PR China.
| |
Collapse
|
2
|
de Amorim IFG, Melo CPDS, Pereira RDA, Cunha SM, Zózimo TRDS, Queiroz FR, Peixoto IDO, Lopes LMS, do Amaral LR, Gomes MDS, Oliveira JA, Cândido EB, Salles PGDO, Braga LDC. Association of a CHEK2 somatic variant with tumor microenvironment calprotectin expression predicts platinum resistance in a small cohort of ovarian carcinoma. PLoS One 2025; 20:e0315487. [PMID: 40146757 PMCID: PMC11949324 DOI: 10.1371/journal.pone.0315487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 11/27/2024] [Indexed: 03/29/2025] Open
Abstract
High-grade serous ovarian cancer (HGSOC) low overall survival rate is often attributed to platinum resistance. Recent studies suggest that the tumor associated-microenvironment (TME) is a determining factor in malignant tumor progression and DNA damage plays a crucial role in this process. Here, we sought to identify platinum resistance biomarkers associating the TME immune profile and the mutational landscape of the homologous repair pathway genes with the HGSOC patients prognosis and response to chemotherapy. Using a decision tree classifier approach, we found that platinum resistant (PR) patients were characterized by the presence of a novel deep intronic variant, the CHEK2 c.319+ 3943A > T, and higher L1 expression (p = 0.016), (100% accuracy). Chek2 protein is an important component of DNA repair and L1, also known as calprotectin, is one component of the neutrophil extracellular traps (NETs) during inflammation, previously suggested as a key contributor to the metastatic process in HGSOC. Also, PD-L2 levels were significantly higher in PR patients positive for this CHEK2 variant (p = 0.048), underscoring the need to explore its potential therapeutic role for this cancer. Our results suggest an interplay between TME and DNA repair variants that results in a multifactorial nature of HGSOC resistance to platinum chemotherapy.
Collapse
Affiliation(s)
- Izabela Ferreira Gontijo de Amorim
- Laboratório de Pesquisa Translacional em Oncologia, Núcleo de Ensino, Pesquisa e Inovação, Instituto Mário Penna, Belo Horizonte, Minas Gerais, Brazil
- Curso de Medicina, Faculdade de Minas-FAMINAS, Belo Horizonte, Minas Gerais, Brazil
| | - Carolina Pereira de Souza Melo
- Laboratório de Pesquisa Translacional em Oncologia, Núcleo de Ensino, Pesquisa e Inovação, Instituto Mário Penna, Belo Horizonte, Minas Gerais, Brazil
| | - Ramon de Alencar Pereira
- Laboratório de Pesquisa Translacional em Oncologia, Núcleo de Ensino, Pesquisa e Inovação, Instituto Mário Penna, Belo Horizonte, Minas Gerais, Brazil
| | - Sidnéa Macioci Cunha
- Hospital Luxemburgo, Instituto Mário Penna, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-graduação em Saúde da Mulher, Departamento de Ginecologia e Obstetrícia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Thalía Rodrigues de Souza Zózimo
- Laboratório de Pesquisa Translacional em Oncologia, Núcleo de Ensino, Pesquisa e Inovação, Instituto Mário Penna, Belo Horizonte, Minas Gerais, Brazil
| | - Fábio Ribeiro Queiroz
- Laboratório de Pesquisa Translacional em Oncologia, Núcleo de Ensino, Pesquisa e Inovação, Instituto Mário Penna, Belo Horizonte, Minas Gerais, Brazil
| | - Iago de Oliveira Peixoto
- Laboratório de Pesquisa Translacional em Oncologia, Núcleo de Ensino, Pesquisa e Inovação, Instituto Mário Penna, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-graduação em Biotecnologia, Fundação Ezequiel Dias-FUNED, Belo Horizonte, Minas Gerais, Brazil
| | - Luciana Maria Silva Lopes
- Programa de Pós-graduação em Biotecnologia, Fundação Ezequiel Dias-FUNED, Belo Horizonte, Minas Gerais, Brazil
| | - Laurence Rodrigues do Amaral
- Laboratório de Bioinformática e Análises Moleculares, Universidade Federal de Uberlândia, Campus Patos de Minas, Uberlândia, Minas Gerais, Brazil
| | - Matheus de Souza Gomes
- Laboratório de Bioinformática e Análises Moleculares, Universidade Federal de Uberlândia, Campus Patos de Minas, Uberlândia, Minas Gerais, Brazil
| | - Juliana Almeida Oliveira
- Curso de Medicina, Faculdade de Minas-FAMINAS, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas à Cirurgia e à Oftalmologia, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Eduardo Batista Cândido
- Curso de Medicina, Faculdade de Minas-FAMINAS, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-graduação em Saúde da Mulher, Departamento de Ginecologia e Obstetrícia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Paulo Guilherme de Oliveira Salles
- Laboratório de Pesquisa Translacional em Oncologia, Núcleo de Ensino, Pesquisa e Inovação, Instituto Mário Penna, Belo Horizonte, Minas Gerais, Brazil
- Hospital Luxemburgo, Instituto Mário Penna, Belo Horizonte, Minas Gerais, Brazil
| | - Letícia da Conceição Braga
- Laboratório de Pesquisa Translacional em Oncologia, Núcleo de Ensino, Pesquisa e Inovação, Instituto Mário Penna, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
3
|
Shi Y, Zhu S, Shan J, Xu Y. Disease-free survival of 15 years after primary surgery in a patient with advanced high-grade serous ovarian cancer: a case report and literature review. Front Oncol 2025; 15:1468196. [PMID: 39931084 PMCID: PMC11807797 DOI: 10.3389/fonc.2025.1468196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 01/06/2025] [Indexed: 02/13/2025] Open
Abstract
Background Ovarian cancer, particularly high-grade serous ovarian cancer (HGSOC), is the most lethal gynecological tumor, with most patients experiencing recurrence within 5 years. Long-term survival in HGSOC patients with advanced stages is exceedingly rare. Case summary We report a case of advanced HGSOC with exceptional long-term recurrence-free survival following initial treatment. In June 2009, the patient underwent suboptimal cytoreductive surgery for stage IIIC ovarian cancer, including total hysterectomy, bilateral salpingo-oophorectomy, omentectomy, appendectomy, and resection of mesenteric and peritoneal lesions. Postoperatively, residual lesions were observed in the mesenteries and para-aortic lymph nodes. Despite unfavorable prognostic factors (advanced stage, aggressive pathology, and incomplete resection), the patient showed remarkable chemosensitivity, remaining recurrence-free for 15 years. Conclusion The factors influencing long-term survival in HGSOC patients are not yet fully understood. We present this rare case to contribute data for further studies on long-term survival in advanced HGSOC.
Collapse
Affiliation(s)
| | | | | | - Yuhong Xu
- Department of Gynecology, Shaoxing People’s Hospital,
Shaoxing, Zhejiang, China
| |
Collapse
|
4
|
Van Kleunen LB, Ahmadian M, Post MD, Wolsky RJ, Rickert C, Jordan KR, Hu J, Richer JK, Brubaker LW, Marjon N, Behbakht K, Sikora MJ, Bitler BG, Clauset A. The Spatial Structure of the Tumor Immune Microenvironment Can Explain and Predict Patient Response in High-Grade Serous Carcinoma. Cancer Immunol Res 2024; 12:1492-1507. [PMID: 39115368 PMCID: PMC11534564 DOI: 10.1158/2326-6066.cir-23-1109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/20/2024] [Accepted: 08/06/2024] [Indexed: 09/18/2024]
Abstract
Ovarian cancer is the deadliest gynecologic malignancy, and therapeutic options and mortality rates over the last three decades have largely not changed. Recent studies indicate that the composition of the tumor immune microenvironment (TIME) influences patient outcomes. To improve spatial understanding of the TIME, we performed multiplexed ion beam imaging on 83 human high-grade serous carcinoma tumor samples, identifying approximately 160,000 cells across 23 cell types. From the 77 of these samples that met inclusion criteria, we generated composition features based on cell type proportions, spatial features based on the distances between cell types, and spatial network features representing cell interactions and cell clustering patterns, which we linked to traditional clinical and IHC variables and patient overall survival (OS) and progression-free survival (PFS) outcomes. Among these features, we found several significant univariate correlations, including B-cell contact with M1 macrophages (OS HR = 0.696; P = 0.011; PFS HR = 0.734; P = 0.039). We then used high-dimensional random forest models to evaluate out-of-sample predictive performance for OS and PFS outcomes and to derive relative feature importance scores for each feature. The top model for predicting low or high PFS used TIME composition and spatial features and achieved an average AUC score of 0.71. The results demonstrate the importance of spatial structure in understanding how the TIME contributes to treatment outcomes. Furthermore, the present study provides a generalizable roadmap for spatial analyses of the TIME in ovarian cancer research.
Collapse
Affiliation(s)
| | - Mansooreh Ahmadian
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Miriam D Post
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | - Rebecca J Wolsky
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | - Christian Rickert
- Department of Immunology and Microbiology, The University of Colorado Anschutz Medical Campus
| | - Kimberly R. Jordan
- Department of Immunology and Microbiology, The University of Colorado Anschutz Medical Campus
| | - Junxiao Hu
- Department of Pediatrics, Cancer Center Biostatistics Core, University of Colorado Anschutz Medical Campus, CO, USA
| | - Jennifer K. Richer
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | | | - Nicole Marjon
- Department of OB/GYN, The University of Colorado Anschutz Medical Campus
| | - Kian Behbakht
- Department of OB/GYN, The University of Colorado Anschutz Medical Campus
| | - Matthew J. Sikora
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | - Benjamin G. Bitler
- Department of OB/GYN, The University of Colorado Anschutz Medical Campus
| | - Aaron Clauset
- Department of Computer Science, University of Colorado, Boulder, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, USA
- Santa Fe Institute, Santa Fe, NM, USA
| |
Collapse
|
5
|
Zhang M, Mo J, Huang W, Bao Y, Luo X, Yuan L. The ovarian cancer-associated microbiome contributes to the tumor's inflammatory microenvironment. Front Cell Infect Microbiol 2024; 14:1440742. [PMID: 39497925 PMCID: PMC11532186 DOI: 10.3389/fcimb.2024.1440742] [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: 06/04/2024] [Accepted: 09/25/2024] [Indexed: 11/07/2024] Open
Abstract
A growing body of research has established a correlation between tumors and persistent chronic inflammatory infiltration. As a primary instigator of inflammation, the majority of microbiomes naturally residing within our bodies engage in a mutually beneficial symbiotic relationship. Nevertheless, alterations in the microbiome's composition or breaches in the normal barrier function can disrupt the internal environment's homeostasis, potentially leading to the development and progression of various diseases, including tumors. The investigation of tumor-related microbiomes has contributed to a deeper understanding of their role in tumorigenesis. This review offers a comprehensive overview of the microbiome alterations and the associated inflammatory changes in ovarian cancer. It may aid in advancing research to elucidate the mechanisms underlying the ovarian cancer-associated microbiome, providing potential theoretical support for the future development of microbiome-targeted antitumor therapies and early screening through convenient methods.
Collapse
Affiliation(s)
- Min Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jiahang Mo
- Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Wu Huang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yiting Bao
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xukai Luo
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Lei Yuan
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Alwosaibai K, Alruwaii ZI, Mashhour M, Almsned FM, Asraf R, Alrsheedy W, Alessa A, Almohanna H, Selwi W, Azam F. Dysgerminomas: germ cell tumors exhibit high expression of PD-L1 and associated with high TILs and good prognosis. Sci Rep 2024; 14:24191. [PMID: 39406772 PMCID: PMC11480429 DOI: 10.1038/s41598-024-74192-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Ovarian germ cell tumors (OVGCTs) account for 28% of all diagnosed ovarian cancers, and malignant germ cell tumors specifically account for approximately 13% of diagnosed ovarian cancers in Saudi Arabia. Although most germ cell tumor patients have a high survival rate, patients who experience tumor recurrence have a poor prognosis and present with more aggressive and chemoresistant tumors. The use of immunotherapeutic agents such as PD-L1/PD-1 inhibitors for OVGCTs remains very limited because few studies have described the immunological characteristics of these tumors. This study is the first to investigate PD-L1 expression in ovarian germ cell tumors and explore the role of PD-L1 expression in tumor microenvironment cells and genetic alterations. A total of 34 ovarian germ cell tumors were collected from pathology archives. The collected tumor tissues included ten dysgerminomas, five yolk sac tumors, five immature teratomas, and one mature teratoma, and the remaining samples were mixed germ cell tumors. The tumors were analyzed using immunohistochemical analysis to determine PD-L1 expression, immune cell infiltration and cancer stem cell populations and their correlation with clinical outcome. Furthermore, the genetic alterations in different subtypes of germ cell tumors were correlated with PD-L1 expression and clinical outcome. Datasets for testicular germ cells (TGCTs) were retrieved from The Cancer Genome Atlas (TCGA) and analyzed using cBioPortal (cbioportal.org) and Gene Expression Profiling Interactive Analysis (GEPIA). Compared with yolk sac tumors, dysgerminomas highly express PD-L1 and are associated with high levels of tumor infiltrating lymphocytes (TILs) and stem cell markers. In addition, compared with PD-L1-negative yolk sac tissue, dysgerminomas/seminomas with high PD-L1 expression are associated with more genetic alterations and a better prognosis. Our findings will contribute to the knowledge about the potential benefits of ovarian cancer immunotherapy in specific subsets of germ cell tumor patients and the risk factors for resistance mediated by tumor microenvironment cells.
Collapse
Affiliation(s)
- Kholoud Alwosaibai
- Biomedical Research Department, Research Center, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia.
| | - Zainab Ibrahim Alruwaii
- Department of Pathology and Lab Medicine, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Miral Mashhour
- Department of Pathology and Lab Medicine, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Fahad M Almsned
- Research Center, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
- School of Systems Biology, George Mason University, Fairfax, USA
| | - Reem Asraf
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Wadha Alrsheedy
- Biomedical Research Department, Research Center, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Ahmed Alessa
- Biomedical Research Department, Research Center, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Hani Almohanna
- Research Center, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Waleed Selwi
- Department of Medical Oncology, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Faisal Azam
- Department of Medical Oncology, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
| |
Collapse
|
7
|
Thompson JJ, McGovern J, Roxburgh CSD, Edwards J, Dolan RD, McMillan DC. The relationship between LDH and GLIM criteria for cancer cachexia: Systematic review and meta-analysis. Crit Rev Oncol Hematol 2024; 199:104378. [PMID: 38754770 DOI: 10.1016/j.critrevonc.2024.104378] [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: 10/12/2023] [Revised: 04/02/2024] [Accepted: 04/30/2024] [Indexed: 05/18/2024] Open
Abstract
INTRODUCTION Cancer cachexia is a clinical condition characterized by recognizable "sickness behaviors" accompanied by loss of lean body tissue. The Global Leadership on Malnutrition (GLIM) has proposed phenotypic (unintentional weight loss, low body mass index and low muscle mass) and aetiologic (reduced food intake and inflammation or disease burden) diagnostic criteria. Recent work has suggested serum lactate dehydrogenase (LDH) might represent a 3rd aetiologic criteria. Little is known of its relationship with GLIM. A systematic review and meta-analysis of their comparative prognostic value and association was performed. METHODS A search of electronic databases (PubMed, Medline, Ovid, Cochrane) up to February 2023 was used to identify studies that compared the prognostic value of LDH and components of the GLIM criteria in cancer. An analysis of the relationship between LDH and the components of GLIM was undertaken where this data was available. RevMan 5.4.1 was used to perform a meta-analysis for each diagnostic criteria that had 3 or more studies which reported hazard ratios with a 95 per cent confidence interval for overall survival (OS). RESULTS A total of 119 studies were reviewed. Advanced lung cancer was the most studied population. Included in the meta-analysis were 6 studies (n=2165) on LDH and weight loss, 17 studies (n=7540) on LDH and low BMI, 5 studies (n=758) on LDH and low muscle mass, 0 studies on LDH and food intake and 93 studies (n=32,190) on LDH and inflammation. There was a significant association between elevated serum LDH and each of low BMI (OR 1.39, 1.09 - 1.77; p=0.008), elevated NLR (OR 2.04, 1.57 - 2.65; p<0.00001) and elevated CRP (OR 2.58, 1.81 - 3.67; p<0.00001). There was no association between elevated serum LDH and low muscle mass. Only one study presented data on the association between LDH and unintentional weight loss. Elevated LDH showed a comparative OS (HR 1.86, 1.57 - 2.07; p<0.00001) to unintentional weight loss (HR 1.57, 1.23 - 1.99; p=0.0002) and had a similar OS (HR 2.00, 1.70 - 2.34; p<0.00001) to low BMI (HR 1.57, 1.29-2.90; p<0.0001). LDH also showed an OS (HR 2.25, 1.76 - 2.87; p<0.00001) congruous with low muscle mass (HR 1.93, 1.14 - 3.27; p=0.01) and again, LDH conferred as poor an OS (HR 1.77, 1.64-1.90; p<0.00001) as elevated NLR (HR 1.61, 1.48 - 1.77; p<0.00001) or CRP (HR 1.55, 1.43 - 1.69; p<0.00001). CONCLUSION Current literature suggests elevated serum LDH is associated with inflammation in cancer (an aetiologic GLIM criterion), however more work is required to establish the relationship between LDH and the phenotypic components of GLIM. Additionally, elevated serum LDH appears to be a comparative prognosticator of overall survival in cancer when compared to the GLIM criteria.
Collapse
Affiliation(s)
- Joshua J Thompson
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK.
| | - Josh McGovern
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
| | - Campbell S D Roxburgh
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
| | - Joanne Edwards
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Ross D Dolan
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
| | - Donald C McMillan
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
| |
Collapse
|
8
|
Van Kleunen L, Ahmadian M, Post MD, Wolsky RJ, Rickert C, Jordan K, Hu J, Richer JK, Marjon NA, Behbakht K, Sikora MJ, Bitler BG, Clauset A. The spatial structure of the tumor immune microenvironment can explain and predict patient response in high-grade serous carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.26.577350. [PMID: 38352574 PMCID: PMC10862769 DOI: 10.1101/2024.01.26.577350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/19/2024]
Abstract
Despite ovarian cancer being the deadliest gynecological malignancy, there has been little change to therapeutic options and mortality rates over the last three decades. Recent studies indicate that the composition of the tumor immune microenvironment (TIME) influences patient outcomes but are limited by a lack of spatial understanding. We performed multiplexed ion beam imaging (MIBI) on 83 human high-grade serous carcinoma tumors - one of the largest protein-based, spatially-intact, single-cell resolution tumor datasets assembled - and used statistical and machine learning approaches to connect features of the TIME spatial organization to patient outcomes. Along with traditional clinical/immunohistochemical attributes and indicators of TIME composition, we found that several features of TIME spatial organization had significant univariate correlations and/or high relative importance in high-dimensional predictive models. The top performing predictive model for patient progression-free survival (PFS) used a combination of TIME composition and spatial features. Results demonstrate the importance of spatial structure in understanding how the TIME contributes to treatment outcomes. Furthermore, the present study provides a generalizable roadmap for spatial analyses of the TIME in ovarian cancer research.
Collapse
Affiliation(s)
- Lucy Van Kleunen
- Department of Computer Science, University of Colorado, Boulder, USA
| | - Mansooreh Ahmadian
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Miriam D Post
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | - Rebecca J Wolsky
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | - Christian Rickert
- Department of Immunology and Microbiology, The University of Colorado Anschutz Medical Campus
| | - Kimberly Jordan
- Department of Immunology and Microbiology, The University of Colorado Anschutz Medical Campus
| | - Junxiao Hu
- Department of Pediatrics, Cancer Center Biostatistics Core, University of Colorado Anschutz Medical Campus, CO, USA
| | - Jennifer K. Richer
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | - Nicole A. Marjon
- Department of OB/GYN, The University of Colorado Anschutz Medical Campus
| | - Kian Behbakht
- Department of OB/GYN, The University of Colorado Anschutz Medical Campus
| | - Matthew J. Sikora
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | - Benjamin G. Bitler
- Department of OB/GYN, The University of Colorado Anschutz Medical Campus
| | - Aaron Clauset
- Department of Computer Science, University of Colorado, Boulder, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, USA
- Santa Fe Institute, Santa Fe, NM, USA
| |
Collapse
|
9
|
Li N, Yu K, Huang D, Zhou H, Zeng D. Identifying a Novel Eight-NK Cell-related Gene Signature for Ovarian Cancer Prognosis Prediction. Curr Med Chem 2024; 31:1578-1594. [PMID: 37650393 DOI: 10.2174/0929867331666230831101847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Ovarian cancer (OVC) is the most common and costly tumor in the world with unfavorable overall survival and prognosis. This study is aimed to explore the prognostic value of natural killer cells related genes for OVC treatment. METHODS RNA-seq and clinical information were acquired from the TCGA-OVC dataset (training dataset) and the GSE51800 dataset (validation dataset). Genes linked to NK cells were obtained from the immPort dataset. Moreover, ConsensusClusterPlus facilitated the screening of molecular subtypes. Following this, the risk model was established by LASSO analysis, and immune infiltration and immunotherapy were then detected by CIBERSORT, ssGSEA, ESTIMATE, and TIDE algorithms. RESULTS Based on 23 NK cell-related genes with prognosis, TCGA-OVC samples were classified into two clusters, namely C1 and C2. Of these, C1 had better survival outcomes as well as enhanced immune infiltration and tumor stem cells. Additionally, it was more suitable for immunotherapy and was also sensitive to traditional chemotherapy drugs. The eight-gene prognosis model was constructed and verified via the GSE51800 dataset. Additionally, a high infiltration level of immune cells was observed in low-risk patients. Low-risk samples also benefited from immunotherapy and chemotherapy drugs. Finally, a nomogram and ROC curves were applied to validate model accuracy. CONCLUSION The present study identified a RiskScore signature, which could stratify patients with different infiltration levels, immunotherapy, and chemotherapy drugs. Our study provided a basis for precisely evaluating OVC therapy and prognosis.
Collapse
Affiliation(s)
- Nan Li
- Reproductive Medicine Center, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, 545001, China
- Liuzhou Institute of Reproduction and Genetics, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, 545001, China
- Guangxi Health Commission Key Laboratory of Birth Cohort Study in Pregnant Women of Advanced Age, Liuzhou, 545001, China
| | - Kai Yu
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Delun Huang
- Department of Physiology, Guangxi University of Chinese Medicine, Nanning, 530004, China
| | - Hui Zhou
- Department of Gynecologic Oncology, Sun Yat-Sen Memorial Hospital of Sun Yatsen University, Guangzhou, 510120, China
| | - Dingyuan Zeng
- Reproductive Medicine Center, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, 545001, China
- Liuzhou Institute of Reproduction and Genetics, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, 545001, China
- Guangxi Health Commission Key Laboratory of Birth Cohort Study in Pregnant Women of Advanced Age, Liuzhou, 545001, China
- The Department of Obstetrics and Gynecology, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, 545001, China
| |
Collapse
|
10
|
Gómez-Valenzuela F, Wichmann I, Suárez F, Kato S, Ossandón E, Hermoso M, Fernández EA, Cuello MA. Cyclooxygenase-2 Blockade Is Crucial to Restore Natural Killer Cell Activity before Anti-CTLA-4 Therapy against High-Grade Serous Ovarian Cancer. Cancers (Basel) 2023; 16:80. [PMID: 38201508 PMCID: PMC10778357 DOI: 10.3390/cancers16010080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Chronic inflammation influences the tumor immune microenvironment (TIME) in high-grade serous ovarian cancer (HGSOC). Specifically, cyclooxygenase-2 (COX-2) overexpression promotes cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) expression. Notably, elevated COX-2 levels in the TIME have been associated with reduced response to anti-CTLA-4 immunotherapy. However, the precise impact of COX-2, encoded by PTGS2, on the immune profile remains unknown. To address this, we performed an integrated bioinformatics analysis using data from the HGSOC cohorts (TCGA-OV, n = 368; Australian cohort AOCS, n = 80; GSE26193, n = 62; and GSE30161, n = 45). Employing Gene Set Variation Analysis (GSVA), MIXTURE and Ecotyper cell deconvolution algorithms, we concluded that COX-2 was linked to immune cell ecosystems associated with shorter survival, cell dysfunction and lower NK cell effector cytotoxicity capacity. Next, we validated these results by characterizing circulating NK cells from HGSOC patients through flow cytometry and cytotoxic assays while undergoing COX-2 and CTLA-4 blockade. The blockade of COX-2 improved the cytotoxic capacity of NK cells against HGSOC cell lines. Our findings underscore the relevance of COX-2 in shaping the TIME and suggest its potential as a prognostic indicator and therapeutic target. Increased COX-2 expression may hamper the effectivity of immunotherapies that require NK cell effector function. These results provide a foundation for experimental validation and clinical trials investigating combined therapies targeting COX-2 and CTLA-4 in HGSOC.
Collapse
Affiliation(s)
- Fernán Gómez-Valenzuela
- Department of Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (F.S.); (S.K.); (E.O.)
| | - Ignacio Wichmann
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 833150, Chile;
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago 833150, Chile
- Division of Oncology, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Felipe Suárez
- Department of Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (F.S.); (S.K.); (E.O.)
| | - Sumie Kato
- Department of Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (F.S.); (S.K.); (E.O.)
| | - Enrique Ossandón
- Department of Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (F.S.); (S.K.); (E.O.)
| | - Marcela Hermoso
- Innate Immunity Laboratory, Immunology Program, Biomedical Sciences Institute, Faculty of Medicine, Universidad de Chile, Santiago 8900085, Chile;
| | - Elmer A. Fernández
- Fundación para el Progreso de la Medicina (CONICET), Córdoba X5000, Argentina;
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba X5000, Argentina
| | - Mauricio A. Cuello
- Department of Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (F.S.); (S.K.); (E.O.)
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago 833150, Chile
- Center for Cancer Prevention and Control (CECAN), Santiago 8330023, Chile
| |
Collapse
|
11
|
Fan Z, Han D, Fan X, Zhao L. Ovarian cancer treatment and natural killer cell-based immunotherapy. Front Immunol 2023; 14:1308143. [PMID: 38187402 PMCID: PMC10768003 DOI: 10.3389/fimmu.2023.1308143] [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: 10/05/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Background Ovarian cancer (OC) is one of the malignant tumors that poses a serious threat to women's health. Natural killer (NK) cells are an integral part of the immune system and have the ability to kill tumor cells directly or participate indirectly in the anti-tumor immune response. In recent years, NK cell-based immunotherapy for OC has shown remarkable potential. However, its mechanisms and effects remain unclear when compared to standard treatment. Methods To explore the value of NK cell-based immunotherapy in the treatment of OC, we conducted a literature review. In comparison to standard treatment, our focus was primarily on the current anti-tumor mechanisms, the clinical effect of NK cells against OC, factors affecting the structure and function of NK cells, and strategies to enhance the effectiveness of NK cells. Results We found that NK cells exert their therapeutic effects in OC through mechanisms such as antibody-dependent cell cytotoxicity, perforin release, and granule enzyme secretion. They also secrete IFN-γ and TNF-α or engage in Fas/FasL and TRAIL/TRAILR pathways, mediating the death of OC cells. In clinical trials, the majority of patients experienced disease stability with mild side effects after receiving NK cell-based immunotherapy, but there is still a lack of high-quality research evidence regarding its clinical effectiveness. OC and prior experience with standard treatments have an effect on NK cells, and it may be considered to maximize NK cell effects through the modulation of the tumor microenvironment or combination with other therapies. Conclusions In this review, we have summarized the current evidence of NK cell applications in the treatment of OC. Furthermore, factors and strategies that influence and enhance the role of NK cell immunotherapy are discussed.
Collapse
Affiliation(s)
- Zhongru Fan
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Dongyu Han
- Department of Obstetrics and Gynecology, Suzhou Hospital, Affiliated Hospital of Meddical School, Nanjing University, Suzhou, China
| | - Xin Fan
- Department of Radiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lin Zhao
- Department of Obstetrics and Gynecology, Suzhou Hospital, Affiliated Hospital of Meddical School, Nanjing University, Suzhou, China
| |
Collapse
|
12
|
Blanc-Durand F, Clemence Wei Xian L, Tan DSP. Targeting the immune microenvironment for ovarian cancer therapy. Front Immunol 2023; 14:1328651. [PMID: 38164130 PMCID: PMC10757966 DOI: 10.3389/fimmu.2023.1328651] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/05/2023] [Indexed: 01/03/2024] Open
Abstract
Ovarian cancer (OC) is an aggressive malignancy characterized by a complex immunosuppressive tumor microenvironment (TME). Immune checkpoint inhibitors have emerged as a breakthrough in cancer therapy by reactivating the antitumor immune response suppressed by tumor cells. However, in the case of OC, these inhibitors have failed to demonstrate significant improvements in patient outcomes, and existing biomarkers have not yet identified promising subgroups. Consequently, there remains a pressing need to understand the interplay between OC tumor cells and their surrounding microenvironment to develop effective immunotherapeutic approaches. This review aims to provide an overview of the OC TME and explore its potential as a therapeutic strategy. Tumor-infiltrating lymphocytes (TILs) are major actors in OC TME. Evidence has been accumulating regarding the spontaneous TILS response against OC antigens. Activated T-helpers secrete a wide range of inflammatory cytokines with a supportive action on cytotoxic T-cells. Simultaneously, mature B-cells are recruited and play a significant antitumor role through opsonization of target antigens and T-cell recruitment. Macrophages also form an important subset of innate immunity (M1-macrophages) while participating in the immune-stimulation context. Finally, OC has shown to engage a significant natural-killer-cells immune response, exerting direct cytotoxicity without prior sensitization. Despite this initial cytotoxicity, OC cells develop various strategies to induce an immune-tolerant state. To this end, multiple immunosuppressive molecules are secreted to impair cytotoxic cells, recruit regulatory cells, alter antigen presentation, and effectively evade immune response. Consequently, OC TME is predominantly infiltrated by immunosuppressive cells such as FOXP3+ regulatory T-cells, M2-polarized macrophages and myeloid-derived suppressor cells. Despite this strong immunosuppressive state, PD-1/PD-L1 inhibitors have failed to improve outcomes. Beyond PD-1/PD-L1, OC expresses multiple other immune checkpoints that contribute to immune evasion, and each representing potential immune targets. Novel immunotherapies are attempting to overcome the immunosuppressive state and induce specific immune responses using antibodies adoptive cell therapy or vaccines. Overall, the OC TME presents both opportunities and obstacles. Immunotherapeutic approaches continue to show promise, and next-generation inhibitors offer exciting opportunities. However, tailoring therapies to individual immune characteristics will be critical for the success of these treatments.
Collapse
Affiliation(s)
- Felix Blanc-Durand
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine and Cancer Science Institute (CSI), National University of Singapore (NUS), Singapore, Singapore
| | - Lai Clemence Wei Xian
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine and Cancer Science Institute (CSI), National University of Singapore (NUS), Singapore, Singapore
| | - David S. P. Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University Centre for Cancer Research (N2CR) and Cancer Science Institute (CSI), National University of Singapore, Singapore, Singapore
| |
Collapse
|
13
|
McKenzie ND, Ahmad S, Awada A, Kuhn TM, Recio FO, Holloway RW. Prognostic features of the tumor microenvironment in high-grade serous ovarian cancer and dietary immunomodulation. Life Sci 2023; 333:122178. [PMID: 37839778 DOI: 10.1016/j.lfs.2023.122178] [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: 09/18/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/17/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is a particularly lethal malignancy that is prognostically influenced by the immune profile of the tumor microenvironment (TME). TME immune profiles have been sub-categorized according to features associated with both survival outcomes as well as response to systemic therapies. Five suggested immune phenotypes have been described and correlated with overall survival outcomes. Phenotypes associated with shorter overall survival rates appear to have prominent immunosuppressive features within their TME. The opportunity to triage patients according to their prognostic TME profile might allow selection of individual patients with poor prognostic features who could most benefit from innovative immunomodulatory treatment strategies. Two potential strategies to indirectly manipulate the TME (and oncologic outcomes) are alteration of the gut microbiome composition and alteration of TME metabolism through dietary interventions. Experimental dietary modifications in humans designed for influencing cancer outcomes are only beginning to be studied in a prospective fashion. Herein we summarize prognostic TME features in HGSOC and potential opportunities for immunomodulation via dietary and gut microbial interventions.
Collapse
Affiliation(s)
- Nathalie D McKenzie
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA.
| | - Sarfraz Ahmad
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA.
| | - Ahmad Awada
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA
| | - Theresa M Kuhn
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA
| | - Fernando O Recio
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA
| | - Robert W Holloway
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA
| |
Collapse
|
14
|
Tassi E, Bergamini A, Wignall J, Sant’Angelo M, Brunetto E, Balestrieri C, Redegalli M, Potenza A, Abbati D, Manfredi F, Cangi MG, Magliacane G, Scalisi F, Ruggiero E, Maffia MC, Trippitelli F, Rabaiotti E, Cioffi R, Bocciolone L, Candotti G, Candiani M, Taccagni G, Schultes B, Doglioni C, Mangili G, Bonini C. Epithelial ovarian cancer is infiltrated by activated effector T cells co-expressing CD39, PD-1, TIM-3, CD137 and interacting with cancer cells and myeloid cells. Front Immunol 2023; 14:1212444. [PMID: 37868997 PMCID: PMC10585363 DOI: 10.3389/fimmu.2023.1212444] [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: 04/26/2023] [Accepted: 09/14/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction Despite predicted efficacy, immunotherapy in epithelial ovarian cancer (EOC) has limited clinical benefit and the prognosis of patients remains poor. There is thus a strong need for better identifying local immune dynamics and immune-suppressive pathways limiting T-cell mediated anti-tumor immunity. Methods In this observational study we analyzed by immunohistochemistry, gene expression profiling and flow cytometry the antigenic landscape and immune composition of 48 EOC specimens, with a focus on tumor-infiltrating lymphocytes (TILs). Results Activated T cells showing features of partial exhaustion with a CD137+CD39+PD-1+TIM-3+CD45RA-CD62L-CD95+ surface profile were exclusively present in EOC specimens but not in corresponding peripheral blood or ascitic fluid, indicating that the tumor microenvironment might sustain this peculiar phenotype. Interestingly, while neoplastic cells expressed several tumor-associated antigens possibly able to stimulate tumor-specific TILs, macrophages provided both co-stimulatory and inhibitory signals and were more abundant in TILs-enriched specimens harboring the CD137+CD39+PD-1+TIM-3+CD45RA-CD62L-CD95+ signature. Conclusion These data demonstrate that EOC is enriched in CD137+CD39+PD-1+TIM-3+CD45RA-CD62L-CD95+ T lymphocytes, a phenotype possibly modulated by antigen recognition on neoplastic cells and by a combination of inhibitory and co-stimulatory signals largely provided by infiltrating myeloid cells. Furthermore, we have identified immunosuppressive pathways potentially hampering local immunity which might be targeted by immunotherapeutic approaches.
Collapse
Affiliation(s)
- Elena Tassi
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, Milano, Italy
- Cell Therapy Immunomonitoring Laboratory (MITiCi), Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alice Bergamini
- Università Vita-Salute San Raffaele, Milan, Italy
- Department of Obstetrics and Gynecology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Jessica Wignall
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Miriam Sant’Angelo
- Department of Surgical Pathology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Emanuela Brunetto
- Department of Surgical Pathology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Chiara Balestrieri
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Miriam Redegalli
- Department of Surgical Pathology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessia Potenza
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Danilo Abbati
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Francesco Manfredi
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, Milano, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Maria Giulia Cangi
- Department of Surgical Pathology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Gilda Magliacane
- Department of Surgical Pathology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Fabiola Scalisi
- Department of Surgical Pathology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Eliana Ruggiero
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Maria Chiara Maffia
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Federica Trippitelli
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Emanuela Rabaiotti
- Department of Obstetrics and Gynecology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Raffaella Cioffi
- Department of Obstetrics and Gynecology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Luca Bocciolone
- Department of Obstetrics and Gynecology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giorgio Candotti
- Department of Obstetrics and Gynecology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Massimo Candiani
- Università Vita-Salute San Raffaele, Milan, Italy
- Department of Obstetrics and Gynecology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Gianluca Taccagni
- Department of Surgical Pathology, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Claudio Doglioni
- Università Vita-Salute San Raffaele, Milan, Italy
- Department of Surgical Pathology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giorgia Mangili
- Department of Obstetrics and Gynecology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Chiara Bonini
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, Milano, Italy
- Cell Therapy Immunomonitoring Laboratory (MITiCi), Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
15
|
Barna AJ, Herold Z, Acs M, Bazsa S, Gajdacsi J, Garay TM, Herold M, Madaras L, Muhl D, Nagy A, Szasz AM, Dank M. High Tumor-Infiltrating Lymphocyte Count Is Associated with Distinct Gene Expression Profile and Longer Patient Survival in Advanced Ovarian Cancer. Int J Mol Sci 2023; 24:13684. [PMID: 37761986 PMCID: PMC10530512 DOI: 10.3390/ijms241813684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/26/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Cancer-related immunity plays a significant role in the outcome of ovarian cancer, but the exact mechanisms are not fully explored. A retrospective, real-life observational study was conducted including 57 advanced ovarian cancer patients. Immunohistochemistry for CD4+, CD8+, and CD45+ was used for assessing tumor-infiltrating immune cells. Furthermore, an immune-related gene expression assay was performed on 12-10 samples from patients with less than and more than 1-year overall survival (OS), respectively. A higher number of CD4+ (p = 0.0028) and CD45+ (p = 0.0221) immune cells within the tumor microenvironment were associated with longer OS of patients. In a multivariate setting, higher CD4+ T cell infiltration predicted longer OS (p = 0.0392). Twenty-three differentially expressed genes-involved in antigen presentation, costimulatory signaling, matrix remodeling, metastasis formation, and myeloid cell activity-were found when comparing the prognostic groups. It was found that tumor-infiltrating immune cell counts are associated with peculiar gene expression patterns and bear prognostic information in ovarian cancer. SOX11 expression emerged and was validated as a predictive marker for OS.
Collapse
Affiliation(s)
- Andras Jozsef Barna
- Department of Obstetrics and Gynecology, Saint Pantaleon Hospital, H-2400 Dunaujvaros, Hungary
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Zoltan Herold
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Miklos Acs
- Department of Surgery, University Hospital, D-93053 Regensburg, Germany
| | - Sandor Bazsa
- Department of Obstetrics and Gynecology, Saint Pantaleon Hospital, H-2400 Dunaujvaros, Hungary
| | - Jozsef Gajdacsi
- Directorate General of Medical Quality Assurance, Semmelweis University, H-1085 Budapest, Hungary
| | - Tamas Marton Garay
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
- Faculty of Information Technology and Bionics, Pazmany Peter Catholic University, H-1083 Budapest, Hungary
| | - Magdolna Herold
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
- Department of Internal Medicine and Hematology, Semmelweis University, H-1088 Budapest, Hungary
| | - Lilla Madaras
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, H-1091 Budapest, Hungary
| | - Dorottya Muhl
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Akos Nagy
- Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary
| | - Attila Marcell Szasz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Magdolna Dank
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| |
Collapse
|
16
|
Lu J, Cai S, Wang F, Wu PY, Pan X, Qiang J, Li H, Zeng M. Development of a prediction model for gross residual in high-grade serous ovarian cancer by combining preoperative assessments of abdominal and pelvic metastases and multiparametric MRI. Acad Radiol 2023; 30:1823-1831. [PMID: 36587996 DOI: 10.1016/j.acra.2022.12.019] [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: 08/10/2022] [Revised: 11/25/2022] [Accepted: 12/11/2022] [Indexed: 01/01/2023]
Abstract
RATIONALE AND OBJECTIVES To preoperatively predict residual tumor (RT) in patients with high-grade serous ovarian carcinoma (HGSOC) via a radiomic-clinical nomogram. METHODS A total of 128 patients with advanced HGSOC were enrolled (training cohort: n=106; validation cohort: n=22). Serum cancer antigen-125 (CA125), serum human epididymis protein 4 (HE-4) level, and neutrophil-to-lymphocyte ratio (NLR) were obtained from the medical records. Metastases in abdomen and pelvis (MAP) of HGSOC patients was evaluated and scored based on preoperative abdominal and pelvic enhanced CT, MRI and/or PET-CT. A volume of interest (VOI) of each tumor was manually contoured along the boundary slice-by-slice. Radiomic features were extracted from the T2-weighted imaging (T2WI), diffusion-weighted imaging (DWI) and apparent diffusion coefficient (ADC) images. Univariate and multivariate analyses were used to determine the independent predictors of RT status. Least absolute shrinkage and selection operator (LASSO) logistic regression was performed to select optimal features and construct radiomic models. A radiomic-clinical nomogram incorporating radiomic signature and clinical parameters was developed and evaluated in training and validation cohorts. RESULTS MAP score (p = 0.002), HE-4 level (p = 0.001) and NLR (p = 0.008) were independent predictors of RT status. The final radiomic-clinical nomogram showed satisfactory prediction performance in training (AUC = 0.936), cross validation (AUC = 0.906) and separate validation cohorts (AUC = 0.900), and fitted well in calibration curves (p > 0.05). Decision curve further confirmed the clinical application value of the nomogram. CONCLUSION The proposed MRI-based radiomic-clinical nomogram achieved excellent preoperative prediction of the RT status in HGSOC.
Collapse
Affiliation(s)
- Jingjing Lu
- Department of Radiology, Zhongshan Hospital, Fudan University, No.180 Fenglin Rd, Shanghai 200032, China; Shanghai Institute of Medical Imaging
| | - Songqi Cai
- Department of Radiology, Zhongshan Hospital, Fudan University, No.180 Fenglin Rd, Shanghai 200032, China; Shanghai Institute of Medical Imaging
| | - Fang Wang
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd. Shanghai, 200232, China
| | - Pu-Yeh Wu
- Department of Research and Development, GE Healthcare, Beijing 100176, China
| | - Xianpan Pan
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd. Shanghai, 200232, China
| | - Jinwei Qiang
- Department of Radiology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai 201508, China
| | - Haiming Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University.
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, No.180 Fenglin Rd, Shanghai 200032, China; Shanghai Institute of Medical Imaging
| |
Collapse
|
17
|
Holloway RW, Mendivil AA, Kendrick JE, Abaid LN, Brown JV, LeBlanc J, McKenzie ND, Mori KM, Ahmad S. Clinical Activity of Olvimulogene Nanivacirepvec-Primed Immunochemotherapy in Heavily Pretreated Patients With Platinum-Resistant or Platinum-Refractory Ovarian Cancer: The Nonrandomized Phase 2 VIRO-15 Clinical Trial. JAMA Oncol 2023; 9:903-908. [PMID: 37227734 PMCID: PMC10214174 DOI: 10.1001/jamaoncol.2023.1007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/16/2023] [Indexed: 05/26/2023]
Abstract
Importance Patients with platinum-resistant or platinum-refractory ovarian cancer (PRROC) have limited therapeutic options, representing a considerable unmet medical need. Objective To assess antitumor activity and safety of intraperitoneal (IP) olvimulogene nanivacirepvec (Olvi-Vec) virotherapy and platinum-based chemotherapy with or without bevacizumab in patients with PRROC. Design, Setting, and Participants This open-label, nonrandomized multisite phase 2 VIRO-15 clinical trial enrolled patients with PRROC with disease progression following their last prior line of therapy from September 2016 to September 2019. Data cutoff was on March 31, 2022, and data were analyzed between April 2022 and September 2022. Interventions Olvi-Vec was administered via a temporary IP dialysis catheter as 2 consecutive daily doses (3 × 109 pfu/d) followed by platinum-doublet chemotherapy with or without bevacizumab. Main Outcomes and Measures Primary outcomes were objective response rate (ORR) via Response Evaluation Criteria in Solid Tumors, version 1.1 (RECIST 1.1) and cancer antigen 125 (CA-125) assay, and progression-free survival (PFS). Secondary outcomes included duration of response (DOR), disease control rate (DCR), safety, and overall survival (OS). Results Twenty-seven heavily pretreated patients with platinum-resistant (n = 14) or platinum-refractory (n = 13) ovarian cancer were enrolled. The median (range) age was 62 (35-78) years. The median (range) prior lines of therapy were 4 (2-9). All patients completed both Olvi-Vec infusions and chemotherapy. Median follow-up duration was 47.0 months (95% CI, 35.9 months to NA). Overall, ORR by RECIST 1.1 was 54% (95% CI, 33%-74%), with a DOR of 7.6 months (95% CI, 3.7-9.6 months). The DCR was 88% (21/24). The ORR by CA-125 was 85% (95% CI, 65%-96%). Median PFS by RECIST 1.1 was 11.0 months (95% CI, 6.7-13.0 months), and the PFS 6-month rate was 77%. Median PFS was 10.0 months (95% CI, 6.4-NA months) in the platinum-resistant group and 11.4 months (95% CI, 4.3-13.2 months) in the platinum-refractory group. The median OS was 15.7 months (95% CI, 12.3-23.8 months) in all patients, with a median OS of 18.5 months (95% CI, 11.3-23.8 months) in the platinum-resistant group and 14.7 months (95% CI, 10.8-33.6 months) in the platinum-refractory group. Most frequent treatment-related adverse events (TRAEs) (any grade, grade 3) were pyrexia (63.0%, 3.7%, respectively) and abdominal pain (51.9%, 7.4%, respectively). There were no grade 4 TRAEs, and no treatment-related discontinuations or deaths. Conclusions and Relevance In this phase 2 nonrandomized clinical trial, Olvi-Vec followed by platinum-based chemotherapy with or without bevacizumab as immunochemotherapy demonstrated promising ORR and PFS with a manageable safety profile in patients with PRROC. These hypothesis-generating results warrant further evaluation in a confirmatory phase 3 trial. Trial Registration ClinicalTrials.gov Identifier: NCT02759588.
Collapse
Affiliation(s)
| | - Alberto A. Mendivil
- Gynecologic Oncology Associates, Newport Beach, California
- Now with Gynecologic Oncology and Complex Pelvic Surgery Program, Hoag Gynecologic Oncology, Newport Beach, California
| | | | - Lisa N. Abaid
- Gynecologic Oncology Associates, Newport Beach, California
- Now with Gynecologic Oncology and Complex Pelvic Surgery Program, Hoag Gynecologic Oncology, Newport Beach, California
| | - John V. Brown
- Gynecologic Oncology Associates, Newport Beach, California
- Now with Gynecologic Oncology and Complex Pelvic Surgery Program, Hoag Gynecologic Oncology, Newport Beach, California
| | | | | | - Kristina M. Mori
- Gynecologic Oncology Associates, Newport Beach, California
- now with Kaiser Permanente, Santa Clara, California
| | | |
Collapse
|
18
|
Adzibolosu N, Alvero AB, Ali-Fehmi R, Gogoi R, Corey L, Tedja R, Chehade H, Gogoi V, Morris R, Anderson M, Vitko J, Lam C, Craig DB, Draghici S, Rutherford T, Mor G. Immunological modifications following chemotherapy are associated with delayed recurrence of ovarian cancer. Front Immunol 2023; 14:1204148. [PMID: 37435088 PMCID: PMC10331425 DOI: 10.3389/fimmu.2023.1204148] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/12/2023] [Indexed: 07/13/2023] Open
Abstract
Introduction Ovarian cancer recurs in most High Grade Serous Ovarian Cancer (HGSOC) patients, including initial responders, after standard of care. To improve patient survival, we need to identify and understand the factors contributing to early or late recurrence and therapeutically target these mechanisms. We hypothesized that in HGSOC, the response to chemotherapy is associated with a specific gene expression signature determined by the tumor microenvironment. In this study, we sought to determine the differences in gene expression and the tumor immune microenvironment between patients who show early recurrence (within 6 months) compared to those who show late recurrence following chemotherapy. Methods Paired tumor samples were obtained before and after Carboplatin and Taxol chemotherapy from 24 patients with HGSOC. Bioinformatic transcriptomic analysis was performed on the tumor samples to determine the gene expression signature associated with differences in recurrence pattern. Gene Ontology and Pathway analysis was performed using AdvaitaBio's iPathwayGuide software. Tumor immune cell fractions were imputed using CIBERSORTx. Results were compared between late recurrence and early recurrence patients, and between paired pre-chemotherapy and post-chemotherapy samples. Results There was no statistically significant difference between early recurrence or late recurrence ovarian tumors pre-chemotherapy. However, chemotherapy induced significant immunological changes in tumors from late recurrence patients but had no impact on tumors from early recurrence patients. The key immunological change induced by chemotherapy in late recurrence patients was the reversal of pro-tumor immune signature. Discussion We report for the first time, the association between immunological modifications in response to chemotherapy and the time of recurrence. Our findings provide novel opportunities to ultimately improve ovarian cancer patient survival.
Collapse
Affiliation(s)
- Nicholas Adzibolosu
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Ayesha B. Alvero
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Rouba Ali-Fehmi
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Radhika Gogoi
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Logan Corey
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Roslyn Tedja
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Hussein Chehade
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Center of Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Vir Gogoi
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Robert Morris
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Matthew Anderson
- Department of Obstetrics and Gynecology, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Julie Vitko
- Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Clarissa Lam
- Department of Gynecologic Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Douglas B. Craig
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, United States
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Sorin Draghici
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, United States
- Advaita Corporation, Ann Arbor, MI, United States
- Division of Information and Intelligent Systems, Directorate for Computer and Information Science and Engineering, National Science Foundation, Alexandria, VA, United States
| | - Thomas Rutherford
- Department of Obstetrics and Gynecology, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Gil Mor
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
19
|
Lei Q, Wang Y, Li J, Wang S, Hu Y, Duan L, Huo Y, Wu Y, Liu H. Increased SEMA6B expression as a potential prognostic and immune cell infiltration biomarker in thyroid cancer patients. Aging (Albany NY) 2023; 15:3572-3585. [PMID: 37155149 PMCID: PMC10449300 DOI: 10.18632/aging.204691] [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: 02/06/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND Even today, thyroid cancer (THCA) remains an important threat to global health. For THCA patients, differentiated thyroid cancer is the most commonly identified pathological subtype, and those diagnosed with papillary thyroid cancer generally have good overall prognosis. For poorly differentiated subtype THCA, patients have aggressive disease course, higher risk of distant organ metastasis and inferior overall prognosis. METHODS RNA-seq data from TCGA and GTEx databases are collected and analyzed via R. The correlation between SEMA6B expression level and pathological as well as clinical parameters of THCA patients was respectively investigated. Gene expression profiling and subsequent functional clustering analysis was the performed utilizing GSEA. The receiver operating characteristic (ROC) curve was utilized to evaluate the diagnostic value of SEMA6B expression. RESULTS Increased SEMA6B expression was characteristic in THCA tumor samples and was associated with specific pathologic and clinical features for TCHA patients. Univariate and multivariate analysis indicated that SEMA6B was independent predictive marker for THCA patients' prognosis. Gene expression profiling and functional clustering analysis suggested that SEMA6B high-expression was related with increased expression of multiple signal pathways and signatures of multiple immune cell infiltration. CONCLUSIONS In this study, through bioinformatic analysis and clinical data investigation, we demonstrated the potential value of SEMA6B as diagnostic and prognostic marker in THCA patient treatment.
Collapse
Affiliation(s)
- Qinghua Lei
- Physical Examination Center, Handan Central Hospital, Congtai, Handan 056008, Hebei Province, China
| | - Yunha Wang
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing 100853, China
| | - Junhua Li
- Physical Examination Center, Handan Central Hospital, Congtai, Handan 056008, Hebei Province, China
| | - Shanshan Wang
- Physical Examination Center, Handan Central Hospital, Congtai, Handan 056008, Hebei Province, China
| | - Yanyan Hu
- Physical Examination Center, Handan Central Hospital, Congtai, Handan 056008, Hebei Province, China
| | - Lihua Duan
- Department of Ultrasound Medicine, Handan Central Hospital, Congtai, Handan 056008, Hebei Province, China
| | - Yanfei Huo
- Physical Examination Center, Handan Central Hospital, Congtai, Handan 056008, Hebei Province, China
| | - Yiping Wu
- Department of Neurology, Handan Central Hospital, Congtai, Handan 056008, Hebei Province, China
| | - Hongzhou Liu
- Department of Endocrinology, First Hospital of Handan City, Congtai, Handan 056002, Hebei Province, China
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing 100853, China
| |
Collapse
|
20
|
Discordance of PD-L1 expression in primary and metastatic ovarian high-grade serous carcinoma and its correlation with CD8 + tumor-infiltrating lymphocytes and patient prognosis. Virchows Arch 2023; 482:755-766. [PMID: 36806916 DOI: 10.1007/s00428-023-03512-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/16/2023] [Accepted: 02/03/2023] [Indexed: 02/23/2023]
Abstract
Differential expression of programmed death-1 ligand (PD-L1) and its clinical significance in primary and metastatic ovarian high-grade serous carcinoma (HGSC) have not been defined. Thus, we investigated the PD-L1 expression of paired ovarian primary and omental metastatic HGSC and its correlation with CD8 + tumor-infiltrating lymphocyte (TILs) and patient survival. A total of 212 cases of ovarian HGSCs with matched primary ovarian and metastatic omental tumors accessioned between 2003 and 2018 were selected for further analysis. Using immunohistochemistry, we evaluated the density of CD8 + TILs and expression of PD-L1 on whole tissue sections. Applying tumor proportion score (TPS, cutoff 1%) and combined positive score (CPS, cutoff 1), the prevalence of PD-L1 expression was similar but with significant discordance in ovarian and omental tumor. Using TPS, patients with PD-L1-positive tumors demonstrated significantly worse recurrence free survival (RFS) and overall survival (OS) than patients with PD-L1-negative tumors. Using CPS, patients with PD-L1-positive ovarian tumors demonstrated significantly worse OS while no significant difference in RFS was found. Patients with PD-L1-positive omental tumors demonstrated significantly worse RFS and OS. Patients with omental PD-L1-positive tumors (TPS) were associated with poorer RFS and OS, while patients with ovarian PD-L1-positive tumors (TPS) were associated with OS not RFS, in COX multivariant analysis. Nonetheless, ovarian and omental high CD8 TILs density was not associated with worse OS in univariant and COX multivariant analysis. PD-L1 expression in ovarian and omental tumor associated with an increased CD8 + TILs density. PD-L1 expression by TPS was better correlated with survival than by CPS, and PD-L1 expression in omental tumors was a stronger prognostic indicator than that in ovarian tumors.
Collapse
|
21
|
Interactions between Platelets and Tumor Microenvironment Components in Ovarian Cancer and Their Implications for Treatment and Clinical Outcomes. Cancers (Basel) 2023; 15:cancers15041282. [PMID: 36831623 PMCID: PMC9953912 DOI: 10.3390/cancers15041282] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Platelets, the primary operatives of hemostasis that contribute to blood coagulation and wound healing after blood vessel injury, are also involved in pathological conditions, including cancer. Malignancy-associated thrombosis is common in ovarian cancer patients and is associated with poor clinical outcomes. Platelets extravasate into the tumor microenvironment in ovarian cancer and interact with cancer cells and non-cancerous elements. Ovarian cancer cells also activate platelets. The communication between activated platelets, cancer cells, and the tumor microenvironment is via various platelet membrane proteins or mediators released through degranulation or the secretion of microvesicles from platelets. These interactions trigger signaling cascades in tumors that promote ovarian cancer progression, metastasis, and neoangiogenesis. This review discusses how interactions between platelets, cancer cells, cancer stem cells, stromal cells, and the extracellular matrix in the tumor microenvironment influence ovarian cancer progression. It also presents novel potential therapeutic approaches toward this gynecological cancer.
Collapse
|
22
|
Falcinelli M, Al-Hity G, Baron S, Mampay M, Allen MC, Samuels M, Jones W, Cilibrasi C, Flaherty RL, Giamas G, Thaker PH, Flint MS. Propranolol reduces IFN-γ driven PD-L1 immunosuppression and improves anti-tumour immunity in ovarian cancer. Brain Behav Immun 2023; 110:1-12. [PMID: 36796704 DOI: 10.1016/j.bbi.2023.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
The immune system plays an important role in controlling epithelial ovarian cancer (EOC). EOC is considered to be a "cold tumour," a tumour that has not triggered a strong response by the immune system. However, tumour infiltrating lymphocytes (TILs) and the expression of programmed cell death ligand (PD-L1) are used as prognostic indicators in EOC. Immunotherapy such as PD-(L)1 inhibitors have shown limited benefit in EOC. Since the immune system is affected by behavioural stress and the beta-adrenergic signalling pathway, this study aimed to explore the impact of propranolol (PRO), a beta-blocker, on anti-tumour immunity in both in vitro and in vivo EOC models. Noradrenaline (NA), an adrenergic agonist, did not directly regulate PD-L1 expression but PD-L1 was significantly upregulated by IFN-γ in EOC cell lines. IFN-γ also increased PD-L1 on extracellular vesicles (EVs) released by ID8 cells. PRO significantly decreased IFN-γ levels in primary immune cells activated ex vivo and showed increased viability of the CD8+ cell population in an EV-immune cell co-incubation. In addition, PRO reverted PD-L1 upregulation and significantly decreased IL-10 levels in an immune-cancer cell co-culture. Chronic behavioural stress increased metastasis in mice while PRO monotherapy and the combo of PRO and PD-(L)1 inhibitor significantly decreased stress-induced metastasis. The combined therapy also reduced tumour weight compared to the cancer control group and induced anti-tumour T-cell responses with significant CD8 expression in tumour tissues. In conclusion, PRO showed a modulation of the cancer immune response by decreasing IFN-γ production and, in turn, IFN-γ-mediated PD-L1 overexpression. The combined therapy of PRO and PD-(L)1 inhibitor decreased metastasis and improved anti-tumour immunity offering a promising new therapy.
Collapse
Affiliation(s)
- M Falcinelli
- University of Brighton, School of Pharmacy & Biosciences, Brighton BN2 4GJ, UK
| | - G Al-Hity
- University of Brighton, School of Pharmacy & Biosciences, Brighton BN2 4GJ, UK
| | - S Baron
- University of Brighton, School of Pharmacy & Biosciences, Brighton BN2 4GJ, UK
| | - M Mampay
- University of Brighton, School of Pharmacy & Biosciences, Brighton BN2 4GJ, UK
| | - M C Allen
- University of Brighton, School of Pharmacy & Biosciences, Brighton BN2 4GJ, UK
| | - M Samuels
- University of Sussex, Department for Biochemistry and Biomedicine, Falmer, Brighton BN1 9QG, UK
| | - W Jones
- University of Sussex, Department for Biochemistry and Biomedicine, Falmer, Brighton BN1 9QG, UK
| | - C Cilibrasi
- University of Sussex, Department for Biochemistry and Biomedicine, Falmer, Brighton BN1 9QG, UK
| | - Renee L Flaherty
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, SW3 6JG London, UK
| | - G Giamas
- University of Sussex, Department for Biochemistry and Biomedicine, Falmer, Brighton BN1 9QG, UK
| | - P H Thaker
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University School of Medicine, St Louis, MO 63110, United States
| | - M S Flint
- University of Brighton, School of Pharmacy & Biosciences, Brighton BN2 4GJ, UK.
| |
Collapse
|
23
|
Body Composition and Metabolic Dysfunction Really Matter for the Achievement of Better Outcomes in High-Grade Serous Ovarian Cancer. Cancers (Basel) 2023; 15:cancers15041156. [PMID: 36831500 PMCID: PMC9953877 DOI: 10.3390/cancers15041156] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/02/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Although obesity-associated metabolic disorders have a negative impact on various cancers, such evidence remains controversial for ovarian cancer. Here, we aimed to evaluate the impact of body composition (BC) and metabolism disorders on outcomes in high-grade serous ovarian cancer (HGSOC). METHODS We analyzed clinical/genomic data from two cohorts (PUC n = 123/TCGA-OV n = 415). BC was estimated using the measurement of adiposity/muscle mass by a CT scan. A list of 425 genes linked to obesity/lipid metabolism was used to cluster patients using non-negative matrix factorization. Differential expression, gene set enrichment analyses, and Ecotyper were performed. Survival curves and Cox-regression models were also built-up. RESULTS We identified four BC types and two clusters that, unlike BMI, effectively correlate with survival. High adiposity and sarcopenia were associated with worse outcomes. We also found that recovery of a normal BC and drug interventions to correct metabolism disorders had a positive impact on outcomes. Additionally, we showed that immune-cell-depleted microenvironments predominate in HGSOC, which was more evident among the BC types and the obesity/lipid metabolism cluster with worse prognosis. CONCLUSIONS We have demonstrated the relevance of BC and metabolism disorders as determinants of outcomes in HGSOC. We have shone a spotlight on the relevance of incorporating corrective measures addressing these disorders to obtain better results.
Collapse
|
24
|
Zhang CL, Jiang XC, Li Y, Pan X, Gao MQ, Chen Y, Pang B. Independent predictive value of blood inflammatory composite markers in ovarian cancer: recent clinical evidence and perspective focusing on NLR and PLR. J Ovarian Res 2023; 16:36. [PMID: 36759864 PMCID: PMC9912515 DOI: 10.1186/s13048-023-01116-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Ovarian cancer (OC) is one of the deadliest malignant tumors affecting women worldwide. The predictive value of some blood inflammatory composite markers in OC has been extensively reported. They can be used for early detection and differential diagnosis of OC and can be used for predicting survival, treatment response, and recurrence in the affected patients. Here, we reviewed the predictive values of composite inflammatory markers based on complete blood count, namely neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), monocyte-to-lymphocyte ratio, and systemic inflammation index and markers based on blood protein, namely C-reactive protein-to-albumin ratio and prognostic nutritional index in OC, with a focus on NLR and PLR. We referred to the clinical studies on these six markers, reviewed the patient population, and summarized the marker cut-off values, significance, and limitations of these studies. All these studies were retrospective and most of them were single-center clinical studies with small sample sizes. We found that the cut-off values of these markers have not been unified, and methods used to determine these values varied among studies. The predictive value of these markers on survival was mainly reflected in the postoperative patients of multiple subtypes of ovarian cancer including epithelial OC, high-grade serous ovarian carcinoma, and ovarian clear cell carcinoma. We focused on NLR and PLR and calculated their pooled hazard ratios. NLR and PLR were reliable in predicting overall and progression-free survivals in patients with OC. Therefore, it is necessary to adjust important confounding factors and conduct a long-term follow-up prospective cohort study to further clarify the cut-off values of NLR and PLR and their clinical applications.
Collapse
Affiliation(s)
- Chuan-long Zhang
- grid.464297.aGuang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053 China
| | - Xiao-chen Jiang
- grid.464297.aGuang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053 China
| | - Yi Li
- grid.464297.aGuang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053 China
| | - Xue Pan
- grid.464297.aGuang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053 China
| | - Meng-qi Gao
- grid.416935.cWangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102 China
| | - Yan Chen
- International Medical Department of Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Bo Pang
- International Medical Department of Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
25
|
Liu L, Wang J, Wu Y, Chen Q, Zhou L, Linghu H, Li Y. A prediction nomogram for suboptimal debulking surgery in patients with serous ovarian carcinoma based on MRI T1 dual-echo imaging and diffusion-weighted imaging. Insights Imaging 2022; 13:204. [PMID: 36575303 PMCID: PMC9794649 DOI: 10.1186/s13244-022-01343-z] [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: 08/09/2022] [Accepted: 12/02/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Serous ovarian carcinoma (SOC) has the highest morbidity and mortality among ovarian carcinoma. Accurate identification of the probability of suboptimal debulking surgery (SDS) is critical. This study aimed to develop a preoperative prediction nomogram of SDS for patients with SOC. METHODS A prediction model was established including 205 patients of SOC from institution A, and 45 patients from institution B were enrolled for external validation. Multivariate logistic regression was used to screen independent predictors and establish a nomogram to predict the occurrence of SDS. RESULTS Multivariate logistic regression demonstrated that the CA-125 level (odds ratio [OR] 8.260, 95% confidence interval [CI] 2.003-43.372), relationship between the sigmoid colon/rectum and ovarian mass (OR 28.701, 95% CI 4.561-286.070), diaphragmatic metastasis (OR 12.369, 95% CI 1.675-274.063), and FIGO stage (OR 32.990, 95% CI 6.623-274.509) were independent predictors for SDS. The area under the curve, concordance index, and 95% CI of the nomogram constructed from the above four factors were 0.951, 0.934, and 0.919-0.982, respectively. The model showed a good fit by the Hosmer-Lemeshow test (training set, p = 0.2475; internal validation set, p = 0.2355; external validation set, p = 0.2707). The external validation proved the reliability of the prediction nomogram. The calibration curve was close to the ideal diagonal line. The decision curve analysis demonstrated a significantly better net benefit. The clinical impact curve indicated good effectiveness in clinical application. CONCLUSION A prediction nomogram for SDS in patients with SOC provides gynecologists with an accurate and effective tool for appropriate management.
Collapse
Affiliation(s)
- Li Liu
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
- Department of Radiology, The People's Hospital of Yubei District of Chongqing City, No. 23 ZhongyangGongyuanBei Road, Yubei District, Chongqing, 401120, China
| | - Jie Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Yan Wu
- Nursing School of Chongqing Medical University, No.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Qiao Chen
- School of Public Health, Chongqing Medical University, No.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Linyi Zhou
- Department of Radiology, Daping Hospital, Army Medical Center, Army Medical University, 10# Changjiangzhilu, Chongqing, 40024, China
| | - Hua Linghu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
26
|
Hudry D, Le Guellec S, Meignan S, Bécourt S, Pasquesoone C, El Hajj H, Martínez-Gómez C, Leblanc É, Narducci F, Ladoire S. Tumor-Infiltrating Lymphocytes (TILs) in Epithelial Ovarian Cancer: Heterogeneity, Prognostic Impact, and Relationship with Immune Checkpoints. Cancers (Basel) 2022; 14:5332. [PMID: 36358750 PMCID: PMC9656626 DOI: 10.3390/cancers14215332] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 08/13/2023] Open
Abstract
Epithelial ovarian cancers (EOC) are often diagnosed at an advanced stage with carcinomatosis and a poor prognosis. First-line treatment is based on a chemotherapy regimen combining a platinum-based drug and a taxane-based drug along with surgery. More than half of the patients will have concern about a recurrence. To improve the outcomes, new therapeutics are needed, and diverse strategies, such as immunotherapy, are currently being tested in EOC. To better understand the global immune contexture in EOC, several studies have been performed to decipher the landscape of tumor-infiltrating lymphocytes (TILs). CD8+ TILs are usually considered effective antitumor immune effectors that immune checkpoint inhibitors can potentially activate to reject tumor cells. To synthesize the knowledge of TILs in EOC, we conducted a review of studies published in MEDLINE or EMBASE in the last 10 years according to the PRISMA guidelines. The description and role of TILs in EOC prognosis are reviewed from the published data. The links between TILs, DNA repair deficiency, and ICs have been studied. Finally, this review describes the role of TILs in future immunotherapy for EOC.
Collapse
Affiliation(s)
- Delphine Hudry
- Inserm, U1192–Protéomique Réponse Inflammatoire Spectrométrie de Masse–PRISM, Lille University, F-59000 Lille, France
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Solenn Le Guellec
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Samuel Meignan
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
- CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille University, F-59000 Lille, France
| | - Stéphanie Bécourt
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Camille Pasquesoone
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Houssein El Hajj
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | | | - Éric Leblanc
- Inserm, U1192–Protéomique Réponse Inflammatoire Spectrométrie de Masse–PRISM, Lille University, F-59000 Lille, France
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Fabrice Narducci
- Inserm, U1192–Protéomique Réponse Inflammatoire Spectrométrie de Masse–PRISM, Lille University, F-59000 Lille, France
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Sylvain Ladoire
- Department of Medical Oncology, Centre Georges-François Leclerc, F-21000 Dijon, France
- INSERM, CRI-866 Faculty of Medicine, F-21000 Dijon, France
| |
Collapse
|
27
|
Ölmez F, Oğlak SC, Ölmez ÖF, Akbayır Ö, Yılmaz E, Akgöl S, Konal M, Seyhan NA, Kinter AK. High expression of CD8 in the tumor microenvironment is associated with PD-1 expression and patient survival in high-grade serous ovarian cancer. Turk J Obstet Gynecol 2022; 19:246-256. [PMID: 36149309 PMCID: PMC9511932 DOI: 10.4274/tjod.galenos.2022.59558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objective: The current study assesses programmed death-1 (PD-1) receptor expression and CD3, CD4, and CD8 tumor-infiltrating lymphocytes (TILs) in high-grade serous ovarian cancer (HGSOC) and associates our results with neoadjuvant chemotherapy history and disease prognosis. Materials and Methods: We included cases diagnosed with primary HGSOC with biopsy or surgical resection materials in this study. The immunoreactivity of CD3, CD4, CD8, and PD1 was assessed immunohistochemically in tumor tissue. We analyzed TILs in two predetermined groups of high and low TIL. The relationships between clinical characteristics, PD-1, and TIL were assessed. by the χ(2) test or Fisher’s Exact test. We used Kaplan-Meier survival analysis and Cox proportional hazards regression model to the connection between survival and the amounts of TIL, and PD1. Results: Univariate analysis demonstrated that optimal debulking (p<0.001), early International Federation of Gynecology and Obstetrics stage (p=0.046), and higher scores of stromal CD8+ TIL expression (p=0.028) in tumor cells were all substantially correlated with longer disease-free survival (DFS), whereas the remaining variables analyzed, including PD-1 positivity, stromal CD3+, and CD4+ TILs, and intraepithelial CD3+, CD4+, and CD8+ TILs, were not correlated with DFS. Also, univariate analysis revealed that optimal debulking (p=0.010), and higher scores of stromal CD8+ TIL expression (p=0.021) in tumor cells were all substantially correlated with longer overall survival (OS). Conclusion: Higher scores of stromal CD8+ TILs are substantially correlated with DFS and OS in univariate analyses, whereas scores of stromal CD3+ and CD4+ TILs, and intraepithelial CD3+, CD4+, and CD8+ TILs are not correlated with DFS and OS in both univariate and multivariate analyses. Also, we found a significant association between PD-1 positivity and the scores of stromal CD3+ TILs and intraepithelial CD8+ TILs. However, no remarkable relationship was revealed between PD-1 positivity and the survival of HGSOC cases.
Collapse
|
28
|
Liu Z, Wu J, Wang X, Ji X. Multivariate logistic regression analysis of the correlation between five biomarkers and ovarian cancer in patients with intermediate-risk: A prospective cross-sectional study. Front Cell Dev Biol 2022; 10:876071. [PMID: 36120557 PMCID: PMC9470860 DOI: 10.3389/fcell.2022.876071] [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: 02/15/2022] [Accepted: 07/18/2022] [Indexed: 11/30/2022] Open
Abstract
Objective: To find potential diagnostic biomarkers for ovarian cancer (OC), a prospective analysis of the expression of five biomarkers in patients with intermediate-risk and their correlation with the occurrence of OC was conducted. Method: A prospective observational study was carried out, patients who underwent surgical treatment with benign or malignant ovarian tumors in our hospital from January 2020 to February 2021 were included in this study, and a total of 263 patients were enrolled. Based on the postoperative pathological results, enrolled patients were divided into ovarian cancer group and benign tumor group (n = 135). The ovarian cancer group was further divided into a mid-stage group (n = 46) and an advanced-stage group (n = 82). The basic information of the three groups of patients was collected, the preoperative imaging data of the patients were collected to assess the lymph node metastasis, the preoperative blood samples were collected to examine cancer antigen 125 (CA125), carbohydrate antigen 19–9 (CA19–9), Neutrophil to lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR), and the postoperative pathological data were sorted and summarized. Result: The average during of disease in the advanced ovarian cancer group was 0.55 ± 0.18 years higher than the benign tumor group (0.43 ± 0.14 years), p < 0.001. In the advanced ovarian cancer group, the ratio of patients with the tumor, node, metastasis (TNM) stage IV (64.63%), with tumor Grade stage II and III (93.90%), and without lymph node metastasis (64.63%) was respectively more than that in the mid-stage group (accordingly 0.00, 36.96, 23.91%) (p < 0.001); The ratio of patients with TNM grade III in the mid-stage group (73.91%) was more than that in the advanced group (35.37%) (p < 0.001). The levels of the five biomarkers: CA19-9, CA125, NLR, PLR, and BDNF were different among the three groups (p < 0.001). Conclusion: CA19-9, CA125, NLR, PLR, BDNF are five biomarkers related to the occurrence of ovarian cancer and are risk factors for it. These five biomarkers and their Combined-Value may be suitable to apply in the diagnosis and the identification of ovarian cancer in patients with intermediate-risk.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Nuclear Medicine, Cangzhou Central Hospital, Cangzhou, China
| | - Jingjing Wu
- Department of Laboratory Medicine, Cangzhou Central Hospital, Cangzhou, China
| | - Xiuli Wang
- Department of Laboratory Medicine, Cangzhou Central Hospital, Cangzhou, China
| | - Xiaoyang Ji
- Department of Obstetrics and Gynecology, Ningjin Hospital of Integrated Traditional Chinese and Western Medicine, Xingtai, China
| |
Collapse
|
29
|
Russell S, Lim F, Peters PN, Wardell SE, Whitaker R, Chang CY, Previs RA, McDonnell DP. Development and Characterization of a Luciferase Labeled, Syngeneic Murine Model of Ovarian Cancer. Cancers (Basel) 2022; 14:cancers14174219. [PMID: 36077756 PMCID: PMC9454869 DOI: 10.3390/cancers14174219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/20/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Despite advances in surgery and targeted therapies, the prognosis for women with high-grade serous ovarian cancer remains poor. Moreover, unlike other cancers, immunotherapy has minimally impacted outcomes in patients with ovarian cancer. Progress in this regard has been hindered by the lack of relevant syngeneic ovarian cancer models to study tumor immunity and evaluate immunotherapies. To address this problem, we developed a luciferase labeled murine model of high-grade serous ovarian cancer, STOSE.M1 luc. We defined its growth characteristics, immune cell repertoire, and response to anti PD-L1 immunotherapy. As with human ovarian cancer, we demonstrated that this model is poorly sensitive to immune checkpoint modulators. By developing the STOSE.M1 luc model, it will be possible to probe the mechanisms underlying resistance to immunotherapies and evaluate new therapeutic approaches to treat ovarian cancer.
Collapse
Affiliation(s)
- Shonagh Russell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
- Correspondence: (S.R.); (D.P.M.); Tel.: +1-919-684-6035 (D.P.M.)
| | - Felicia Lim
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Pamela N. Peters
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Suzanne E. Wardell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Regina Whitaker
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Rebecca A. Previs
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Donald P. McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
- Correspondence: (S.R.); (D.P.M.); Tel.: +1-919-684-6035 (D.P.M.)
| |
Collapse
|
30
|
Prieto-Potin I, Idrovo F, Suárez-Gauthier A, Díaz-Blázquez M, Astilleros-Blanco de Córdova L, Chamizo C, Zazo S, Carvajal N, López-Sánchez A, Pérez-Buira S, Aúz-Alexandre CL, Manso R, Plaza-Sánchez J, de Lucas-López V, Pérez-González N, Martín-Valle S, Cristóbal I, Casado V, García-Foncillas J, Rojo F. Comprehensive Approach to Genomic and Immune Profiling: Insights of a Real-World Experience in Gynecological Tumors. Diagnostics (Basel) 2022; 12:diagnostics12081903. [PMID: 36010253 PMCID: PMC9406465 DOI: 10.3390/diagnostics12081903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022] Open
Abstract
Gynecological cancer accounts for an elevated incidence worldwide requiring responsiveness regarding its care. The comprehensive genomic approach agrees with the classification of certain tumor types. We evaluated 49 patients with gynecological tumors undergoing high-throughput sequencing to explore whether identifying alterations in cancer-associated genes could characterize concrete histological subtypes. We performed immune examination and analyzed subsequent clinical impact. We found 220 genomic aberrations mostly distributed as single nucleotide variants (SNV, 77%). Only 3% were classified as variants of strong clinical significance in BRCA1 and BRCA2 of ovarian high-grade serous (HGSC) and uterine endometrioid carcinoma. TP53 and BRCA1 occurred in 72% and 28% of HGSC. Cervical squamous cell carcinoma was entirely HPV-associated and mutations occurred in PIK3CA (60%), as well as in uterine serous carcinoma (80%). Alterations were seen in PTEN (71%) and PIK3CA (60%) of uterine endometrioid carcinoma. Elevated programmed death-ligand 1 (PD-L1) was associated with high TILs. Either PD-L1 augmented in deficient mis-matched repair (MMR) proteins or POLE mutated cases when compared to a proficient MMR state. An 18% received genotype-guided therapy and a 4% immunotherapy. The description of tumor subtypes is plausible through high-throughput sequencing by recognizing clinically relevant alterations. Additional concomitant assessment of immune biomarkers identifies candidates for immunotherapy.
Collapse
Affiliation(s)
- Iván Prieto-Potin
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Franklin Idrovo
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Ana Suárez-Gauthier
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - María Díaz-Blázquez
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | | | - Cristina Chamizo
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Sandra Zazo
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Nerea Carvajal
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Almudena López-Sánchez
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Sandra Pérez-Buira
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Carmen Laura Aúz-Alexandre
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Rebeca Manso
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Jenifer Plaza-Sánchez
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Virginia de Lucas-López
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Nuria Pérez-González
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Sara Martín-Valle
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Ion Cristóbal
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
- Translational Oncology Division, Oncohealth Institute, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Victoria Casado
- Translational Oncology Division, Oncohealth Institute, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Jesús García-Foncillas
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
- Translational Oncology Division, Oncohealth Institute, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
| | - Federico Rojo
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, 28040 Madrid, Spain
- Correspondence:
| |
Collapse
|
31
|
Qin M, Chen G, Hou J, Wang L, Wang Q, Wang L, Jiang D, Hu Y, Xie B, Chen J, Wei H, Xu G. Tumor-infiltrating lymphocyte: features and prognosis of lymphocytes infiltration on colorectal cancer. Bioengineered 2022; 13:14872-14888. [PMID: 36633318 PMCID: PMC9995135 DOI: 10.1080/21655979.2022.2162660] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) are vital elements of the tumor microenvironment (TME), and the anti-tumor activity of TILs on colorectal cancer (CRC) has been a topic of concern. However, the characteristics and prognosis of the various types of lymphocyte infiltration in CRC have not been fully explained. Our study aimed to identify distinct features and prognosis of TILs. We integrated multiple-cohort databases to illustrate the features, proportions, and prognosis of TILs on CRC. We found that macrophages were significantly enriched in CRC. When we used the scRNA-seq database to further evaluate the proportion of TILs, we noticed markedly higher numbers of CD4 + T cell, B cell, and CD8 + T cell in four Gene Expression Omnibus Series (GSE) CRC cohorts. Interestingly, we found that the infiltrating level of TIL subgroups from highest to lowest is always dendritic cells, CD8 + T cells, CD4 + T cells, neutrophils, B cells, and macrophages; the proportion of infiltration is largely constant regardless of mutations in specific genes or somatic copy number variation (sCNV). In addition, the data corroborated that CD4+ TILs and CD8+ TILs have certain application values in the prognosis of CRCs, and age negatively related to CD8+ TILs and B plasma infiltration. Finally, patients with CRC who are older than 70 years have a better response to immune-checkpoint blockade.
Collapse
Affiliation(s)
- Miao Qin
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Gang Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jinxia Hou
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Li Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Qunfeng Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Lina Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Dan Jiang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Institute of Clinical Laboratory, Guangdong Medical University, Dongguan, China
| | - Ye Hu
- Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bei Xie
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jing Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Hulai Wei
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Guangxian Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Institute of Clinical Laboratory, Guangdong Medical University, Dongguan, China
| |
Collapse
|
32
|
Han J, Zhao L, Wu J, Diao Y, Guo Q, Yang J, Luo Y. Role of CD4 +T, CD8 +T Cells, and CD4 +T/CD8 +T Cell Ratio in Gastric Cancer and Its Clinical Significance. Appl Bionics Biomech 2022; 2022:1094607. [PMID: 35535325 PMCID: PMC9078800 DOI: 10.1155/2022/1094607] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 11/18/2022] Open
Abstract
Objective To study the expression and clinical importance of CD4+T, CD8+T cells, and CD4+T/CD8+T cell percentage in gastric cancer (GC) patients. Methods The blood count of CD4+T and CD8+T lymphocytes was ascertained via flow cytometry before surgery in 93 GC patients undergoing gastrectomy. The CD4+T, CD8+T, and Foxp3+T lymphocytes in cancerous and normal adjacent tissues and the presence of PD-L1 in cancerous tissues were detected via immunohistochemistry. The link between the permeation of CD4+T, CD8+T lymphocytes in venous blood, and cancer and normal adjacent tissues was analyzed. Results Lauren histotype, TNM stage, lymphatic/nervous invasion, and NLR level were all considerably associated with peripheral CD4+T and CD8+T cell levels, whereas CD8+T lymphocytes were also associated with vascular invasion (p < 0.05). The CD4+T lymphocyte counts, CD4+T, and CD8+T cell percentage in GC tissues were found to have been decreased when compared to normal adjacent tissues, whereas the CD8+T and Foxp3+T lymphocyte count was higher in GC tissues (p < 0.05). According to a Spearman analysis, the CD4+T and CD8+T cell counts in tumor tissues were positively related to the Foxp3+T lymphocyte count (p < 0.05). Greater peripheral CD4+T lymphocyte counts and increased level of CD4+T/CD8+T percentage corresponded with greater CD4+T cell levels and increased CD4+T/CD8+T quantity in normal adjacent tissues. Higher levels of peripheral CD8+T cells corresponded with higher quantities of CD8+T cells in cancer tissues. A reduced CD4+T lymphocyte count, together with a reduced CD4+T/CD8+T percentage in venous blood, was consistent with a diminished CD4+T cell count along with a reduced CD4+T/CD8+T lymphocyte ratio in cancer and normal adjacent tissues. Conclusion The peripheral quantity of CD4+T and CD8+T lymphocytes in GC patients can partly reflect the infiltrating state of these lymphocytes in cancer and normal adjacent tissues and can preliminarily predict immunotherapy response to a certain extent.
Collapse
Affiliation(s)
- Jingqi Han
- Department of Pathology, Affiliated Hospital of Qinghai University, Xining, China
| | - Linglin Zhao
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- Department of Emergency, Qinghai Provincial People's Hospital, Xining, China
| | - Jianying Wu
- Department of Virology, Qinghai Center for Disease Prevention and Control, Xining, China
| | - Yinzhuo Diao
- Department of Oncology, Qinghai Province Cancer Hospital, Xining, China
| | - Qijing Guo
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, China
| | - Jie Yang
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, China
| | - Yushuang Luo
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, China
| |
Collapse
|
33
|
Ok Atılgan A, Yılmaz Akçay E, Özen Ö, Haberal Reyhan AN, Ayhan A. The Overexpression of Programmed Death-Ligand 2 in Uterine Adenosarcoma: Correlation with High-Grade Morphology, Mutant Type TP53 Expression and Clinical Outcomes. Int J Surg Pathol 2022; 31:352-364. [PMID: 35466759 DOI: 10.1177/10668969221095189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immunotherapy involving the programmed death-1 (PD-1)/the programmed death-ligand (PD-1/PD-L) blockade is an understudied tumor therapy approach in cases of adenosarcoma. PD-L1 and PD-L2, and tumor protein p53 (p53) were examined in 20 uterine adenosarcoma cases, and tumor-infiltrating lymphocytes and tumor-associated macrophages were counted in tumor tissue using immunohistochemistry. While CPS PD-L1 positivity with 1% and 10% cut-off values was observed in 40% and 10% of tumors, respectively, CPS PD-L2 positivity with 1%, 10% and 50% cut-off values was observed in 100%, 85% and 50% of the tumors, respectively. The CPS PD-L2 positivity with a 50% cut-off value was positively correlated with tumor grade and the presence of sarcomatous overgrowth and lymphovascular invasion (LVI) (p = 0.025, p = 0.025, and p = 0.025, respectively). Nine of 11 high-grade adenosarcomas and none of the low-grade adenosarcomas showed mutant type p53 expression (p = 0.000). However, PD-L1 expression and tumor-infiltrating immune cells did not correlate with clinicopathological parameters. The CPS PD-L2 positivity with a 50% cut-off value was also positively correlated with mutant type p53 expression (p = 0.024) and tumor-associated macrophages density (p = 0.024). The CPS PD-L2 positivity with a 50% cut-off value and mutant type p53 expression were associated with shorter disease-free survival and shorter overall survival. The high density of tumor-associated macrophages and low density of tumor-infiltrating lymphocytes were also associated with shorter disease-free survival and overall survival (p < 0.05).These results suggested that the CPS PD-L2 positivity with a 50% cut-off value, p53 mutation and tumor microenvironment played an essential role in the progression of uterine adenosarcomas.
Collapse
Affiliation(s)
- Alev Ok Atılgan
- Department of Pathology, Faculty of Medicine, Baskent University, Bahcelievler, Ankara, Turkey
| | - Eda Yılmaz Akçay
- Department of Pathology, Faculty of Medicine, Baskent University, Bahcelievler, Ankara, Turkey
| | - Özlem Özen
- Department of Pathology, Faculty of Medicine, Baskent University, Bahcelievler, Ankara, Turkey
| | - A. Nihan Haberal Reyhan
- Department of Pathology, Faculty of Medicine, Baskent University, Bahcelievler, Ankara, Turkey
| | - Ali Ayhan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Baskent University, Faculty of Medicine, Bahcelievler, Ankara, Turkey
| |
Collapse
|
34
|
Shen X, Gu X, Ma R, Li X, Wang J. Identification of the Immune Signatures for Ovarian Cancer Based on the Tumor Immune Microenvironment Genes. Front Cell Dev Biol 2022; 10:772701. [PMID: 35372348 PMCID: PMC8974491 DOI: 10.3389/fcell.2022.772701] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 03/02/2022] [Indexed: 11/21/2022] Open
Abstract
Ovarian cancer (OV) is a deadly gynecological cancer. The tumor immune microenvironment (TIME) plays a pivotal role in OV development. However, the TIME of OV is not fully known. Therefore, we aimed to provide a comprehensive network of the TIME in OV. Gene expression data and clinical information from OV patients were obtained from the Cancer Genome Atlas Program (TCGA) database. Non-negative Matrix Factorization, NMFConsensus, and nearest template prediction algorithms were used to perform molecular clustering. The biological functions of differentially expressed genes (DEGs) were identified using Metascape, gene set enrichment analysis (GSEA), gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. The copy number variations (CNVs), single nucleotide polymorphisms (SNPs) and tumor mutation burden were analyzed using Gistic 2.0, R package maftools, and TCGA mutations, respectively. Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data and CIBERSORT were utilized to elucidate the TIME. Moreover, external data from the International Cancer Genome Consortium (ICGC) and ArrayExpress databases were used to validate the signature. All 361 samples from the TCGA OV dataset were classified into Immune Class and non-Immune Class with immune signatures. By comparing the two classes, we identified 740 DEGs that accumulated in immune-related, cancer-related, inflammation-related biological functions and pathways. There were significant differences in the CNVs between the Immune and non-Immune Classes. The Immune Class was further divided into immune-activated and immune-suppressed subtypes. There was no significant difference in the top 20 genes in somatic SNPs among the three groups. In addition, the immune-activated subtype had significantly increased proportions of CD4 memory resting T cells, T cells, M1 macrophages, and M2 macrophages than the other two groups. The qRT-PCR results indicated that the mRNA expression levels of RYR2, FAT3, MDN1 and RYR1 were significantly down-regulated in OV compared with normal tissues. Moreover, the signatures of the TIME were validated using ICGC cohort and the ArrayExpress cohort. Our study clustered the OV patients into an immune-activated subtype, immune-suppressed subtype, and non-Immune Class and provided potential clues for further research on the molecular mechanisms and immunotherapy strategies of OV.
Collapse
Affiliation(s)
- Xiaoyan Shen
- Department of Gynecology, Peking University People’s Hospital, Beijing, China
| | - Xiao Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ruiqiong Ma
- Department of Gynecology, Peking University People’s Hospital, Beijing, China
| | - Xiaoping Li
- Department of Gynecology, Peking University People’s Hospital, Beijing, China
| | - Jianliu Wang
- Department of Gynecology, Peking University People’s Hospital, Beijing, China
- *Correspondence: Jianliu Wang,
| |
Collapse
|
35
|
Launonen IM, Lyytikäinen N, Casado J, Anttila EA, Szabó A, Haltia UM, Jacobson CA, Lin JR, Maliga Z, Howitt BE, Strickland KC, Santagata S, Elias K, D'Andrea AD, Konstantinopoulos PA, Sorger PK, Färkkilä A. Single-cell tumor-immune microenvironment of BRCA1/2 mutated high-grade serous ovarian cancer. Nat Commun 2022; 13:835. [PMID: 35149709 PMCID: PMC8837628 DOI: 10.1038/s41467-022-28389-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/14/2022] [Indexed: 11/12/2022] Open
Abstract
The majority of high-grade serous ovarian cancers (HGSCs) are deficient in homologous recombination (HR) DNA repair, most commonly due to mutations or hypermethylation of the BRCA1/2 genes. We aimed to discover how BRCA1/2 mutations shape the cellular phenotypes and spatial interactions of the tumor microenvironment. Using a highly multiplex immunofluorescence and image analysis we generate spatial proteomic data for 21 markers in 124,623 single cells from 112 tumor cores originating from 31 tumors with BRCA1/2 mutation (BRCA1/2mut), and from 13 tumors without alterations in HR genes. We identify a phenotypically distinct tumor microenvironment in the BRCA1/2mut tumors with evidence of increased immunosurveillance. Importantly, we report a prognostic role of a proliferative tumor-cell subpopulation, which associates with enhanced spatial tumor-immune interactions by CD8+ and CD4 + T-cells in the BRCA1/2mut tumors. The single-cell spatial landscapes indicate distinct patterns of spatial immunosurveillance with the potential to improve immunotherapeutic strategies and patient stratification in HGSC.
Collapse
Affiliation(s)
- I-M Launonen
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - N Lyytikäinen
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - J Casado
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - E A Anttila
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - A Szabó
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - U-M Haltia
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
| | - C A Jacobson
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - J R Lin
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Z Maliga
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - B E Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - K C Strickland
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - S Santagata
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
| | - K Elias
- Department of Obstetrics and Gynecology and Reproductive Biology, Brigham and Women's Hospital, Boston, MA, USA
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - A D D'Andrea
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - P A Konstantinopoulos
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - P K Sorger
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
| | - A Färkkilä
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland.
- Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland.
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA.
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland.
| |
Collapse
|
36
|
Iwahashi H, Miyamoto M, Ito T, Suminokura J, Hada T, Ishibashi H, Kakimoto S, Matsuura H, Suzuki R, Minabe S, Matsukuma S, Tsuda H, Takano M. Clinical significance of CD8-positive lymphocytes on tumor cell clusters of ascites cell block in ovarian high-grade serous carcinoma. Cancer Med 2022; 11:2085-2095. [PMID: 35137571 PMCID: PMC9119359 DOI: 10.1002/cam4.4592] [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: 11/22/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 11/11/2022] Open
Abstract
Background The clinical significance of CD8‐positive (CD8+) lymphocytes on tumor cell clusters of ascites cell blocks in patients with ovarian high‐grade serous carcinoma (HGSC) was investigated. Methods Among HGSC patients who underwent surgery from January 2014 to December 2019, 38 patients with ascites cell block were selected. Using these cell blocks and primary ovarian tumor tissue, the presence of CD8+ lymphocytes and the expression of PD‐L1 were examined immunohistochemically. Tumor cell clusters were defined as cell clumps consisting of more than 10 malignant cells in cell block. Cases with at least one CD8+ lymphocyte in tumor cell cluster were defined as positive CD8+ lymphocytes (Group A); others were defined as negative CD8+ lymphocytes (Group B). The tumor tissue CD8+ lymphocytes were counted mechanically. Clinicopathological features were retrospectively compared between the two groups. Results In total, 38 cases were identified: 25 (65.8%) in Group A and 13 (34.2%) in Group B. More cases in Group A were positive for CD4 (p < 0.01), PD‐L1 (p = 0.02), FoxP3 (p = 0.02) and had a higher number of CD8+ lymphocytes in the tissue (p = 0.03). Patients in Group A had better progression‐free survival (p < 0.01) and overall survival (p = 0.04). In multivariate analysis, Group A was an independent prognostic factor for both progression‐free survival (hazard ratio, 0.24; p < 0.01) and overall survival (hazard ratio, 0.21; p = 0.03). Conclusion The presence of CD8+ lymphocytes in tumor cell clusters of ascites was associated with the status of immune reaction in the tissue and prognosis in patients with HGSC and might be useful information of the immune‐associated therapy.
Collapse
Affiliation(s)
- Hideki Iwahashi
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Morikazu Miyamoto
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Tsubasa Ito
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Jin Suminokura
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Taira Hada
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Hiroki Ishibashi
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Soichiro Kakimoto
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Hiroko Matsuura
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Rie Suzuki
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Shinya Minabe
- Department of Laboratory Medicine, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Susumu Matsukuma
- Department of Laboratory Medicine, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Hitoshi Tsuda
- Department of Basic Pathology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Masashi Takano
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| |
Collapse
|
37
|
Li Y, Gong X, Hu T, Chen Y. Two novel prognostic models for ovarian cancer respectively based on ferroptosis and necroptosis. BMC Cancer 2022; 22:74. [PMID: 35039008 PMCID: PMC8764839 DOI: 10.1186/s12885-021-09166-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 12/30/2021] [Indexed: 12/01/2022] Open
Abstract
Background Platinum-resistant cases account for 25% of ovarian cancer patients. Our aim was to construct two novel prognostic models based on gene expression data respectively from ferroptosis and necroptosis, for predicting the prognosis of advanced ovarian cancer patients with platinum treatment. Methods According to the different overall survivals, we screened differentially expressed genes (DEGs) from 85 ferroptosis-related and 159 necroptosis-related gene expression data in the GSE32062 cohort, to establish two ovarian cancer prognostic models based on calculating risk factors of DEGs, and log-rank test was used for statistical significance test of survival data. Subsequently, we validated the two models in the GSE26712 cohort and the GSE17260 cohort. In addition, we took gene enrichment and microenvironment analyses respectively using limma package and GSVA software to compare the differences between high- and low-risk ovarian cancer patients. Results We constructed two ovarian cancer prognostic models: a ferroptosis-related model based on eight-gene expression signature and a necroptosis-related model based on ten-gene expression signature. The two models performed well in the GSE26712 cohort, but the performance of necroptosis-related model was not well in the GSE17260 cohort. Gene enrichment and microenvironment analyses indicated that the main differences between high- and low- risk ovarian cancer patients occurred in the immune-related indexes, including the specific immune cells abundance and overall immune indexes. Conclusion In this study, ovarian cancer prognostic models based on ferroptosis and necroptosis have been preliminarily validated in predicting prognosis of advanced patients treated with platinum drugs. And the risk score calculated by these two models reflected immune microenvironment. Future work is needed to find out other gene signatures and clinical characteristics to affect the accuracy and applicability of the two ovarian cancer prognostic models. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-09166-9.
Collapse
Affiliation(s)
- Yang Li
- Department of Obstetrics and Gynecology, Tianjin Hospital, Tianjin, 300211, China
| | - Xiaojin Gong
- Department of Obstetrics and Gynecology, Tianjin Hospital, Tianjin, 300211, China
| | - Tongxiu Hu
- Department of Obstetrics and Gynecology, Tianjin Hospital, Tianjin, 300211, China
| | - Yurong Chen
- Department of Oncology, Zhuji People's Hospital of Zhejiang Province, Zhuji, 311800, Zhejiang, China.
| |
Collapse
|
38
|
Nishida S, Morimoto S, Oji Y, Morita S, Shirakata T, Enomoto T, Tsuboi A, Ueda Y, Yoshino K, Shouq A, Kanegae M, Ohno S, Fujiki F, Nakajima H, Nakae Y, Nakata J, Hosen N, Kumanogoh A, Oka Y, Kimura T, Sugiyama H. Cellular and Humoral Immune Responses Induced by an HLA Class I-restricted Peptide Cancer Vaccine Targeting WT1 Are Associated With Favorable Clinical Outcomes in Advanced Ovarian Cancer. J Immunother 2022; 45:56-66. [PMID: 34874330 PMCID: PMC8654282 DOI: 10.1097/cji.0000000000000405] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/22/2021] [Indexed: 11/25/2022]
Abstract
The HLA-A*24:02-restricted peptide vaccine targeting Wilms' tumor 1 (WT1) (WT1 vaccine) is a promising therapeutic strategy for ovarian cancer; however, its efficacy varies among patients. In this study, we analyzed WT1-specific immune responses in patients with advanced or recurrent ovarian cancer that was refractory to standard chemotherapies and their associations with clinical outcomes. In 25 patients, the WT1 vaccine was administered subcutaneously weekly for 3 months and biweekly thereafter until disease progression or severe adverse events. We assessed Wilms' tumor 1-specific cytotoxic T lymphocytes (WT1-CTLs) and Wilms' tumor 1 peptide-specific immunoglobulin G (WT1235-IgG). After vaccination, the percentage of tetramer high-avidity population of WT1-CTLs among CD8+ T lymphocytes (%tet-hi WT1-CTL) and the WT1235-IgG titer increased significantly, although the values were extremely low or below the limit of detection before vaccination (%tet-hi WT1-CTL: 0.003%-0.103%.; WT1235-IgG: <0.05-0.077 U/mL). Patients who had %tet-hi WT1-CTL of ≥0.25% (n=6) or WT1235-IgG of ≥0.10 U/mL (n=12) had a significantly longer progression-free survival than those of patients in the other groups. In addition, an increase in WT1235-IgG corresponded to a significantly longer progression-free survival (P=0.0496). In patients with systemic inflammation, as evidenced by elevated C-reactive protein levels, the induction of tet-hi WT1-CTL or WT1235-IgG was insufficient. Decreased serum albumin levels, multiple tumor lesions, poor performance status, and excess ascites negatively influenced the clinical effectiveness of the WT1 vaccine. In conclusion, the WT1 vaccine induced antigen-specific cellular and humoral immunity in patients with refractory ovarian cancer. Both %tet-hi WT1-CTL and WT1235-IgG levels are prognostic markers for the WT1 vaccine.
Collapse
Affiliation(s)
| | | | | | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto
| | | | - Takayuki Enomoto
- Obstetrics and Gynecology, Osaka University Graduates School of Medicine
- Department of Obstetrics and Gynecology, Niigata University Medical School, Niigata
| | | | - Yutaka Ueda
- Obstetrics and Gynecology, Osaka University Graduates School of Medicine
| | - Kiyoshi Yoshino
- Obstetrics and Gynecology, Osaka University Graduates School of Medicine
- Department of Obstetrics and Gynecology, University of Occupational and Environmental Health, Kita-Kyushu, Fukuoka Prefecture
| | | | | | - Satoshi Ohno
- Cancer Immunotherapy
- Clinical Research Support Center, Shimane University Faculty of Medicine, Izumo, Shimane Prefecture, Japan
| | | | | | - Yoshiki Nakae
- Departments of Respiratory Medicine and Clinical Immunology
| | | | | | - Atsushi Kumanogoh
- Departments of Respiratory Medicine and Clinical Immunology
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka Prefecture
| | - Yoshihiro Oka
- Cancer Stem Cell Biology
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University
| | - Tadashi Kimura
- Obstetrics and Gynecology, Osaka University Graduates School of Medicine
| | | |
Collapse
|
39
|
Zhang T, Liu Q, Zhu Y, Huang Y, Qin J, Wu X, Zhang S. Lymphocyte and macrophage infiltration in omental metastases indicates poor prognosis in advance stage epithelial ovarian cancer. J Int Med Res 2021; 49:3000605211066245. [PMID: 34939862 PMCID: PMC8721736 DOI: 10.1177/03000605211066245] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective To investigate the prognostic value of immune cells within omental metastases originating from advanced epithelial ovarian cancer (EOC). Methods We performed immunohistochemical analysis to determine the levels of CD4+/CD8+ tumor-infiltrating lymphocytes (TILs) and CD68+ tumor-associated microphages (TAMs) in omental specimens from 100 patients with advanced EOC. Significant prognostic factors, including immune cells and clinical parameters, were assessed by Kaplan–Meier survival analysis and Cox models. Results Cox regression analysis showed that elevated levels of CD68+ TAMs and intra-islet CD4+ TILs in omental metastases were the main risk factors associated with worse survival outcomes for advanced EOC. Moreover, the survival analysis of relationships between omental immune cells and favorable clinical predictors revealed additional prognostic stratification information. Conclusion Omental immune cells (TAMs and TILs) provide alternative prognostic factors in advanced EOC. In contrast to markers of the EOC tumor microenvironment at the primary site, elevated CD68+ TAMs and intra-islet CD4+ TILs in omental metastases serve as negative prognostic markers in advanced EOC and imply an unfavorable outcome.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Oncology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qin Liu
- Department of Pathology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yingfan Zhu
- Department of Gynecology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yizhou Huang
- Department of Gynecology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiale Qin
- Department of Oncology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaodong Wu
- Department of Oncology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Songfa Zhang
- Department of Oncology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
40
|
Wang JJ, Siu MKY, Jiang YX, Leung THY, Chan DW, Wang HG, Ngan HYS, Chan KKL. A Combination of Glutaminase Inhibitor 968 and PD-L1 Blockade Boosts the Immune Response against Ovarian Cancer. Biomolecules 2021; 11:biom11121749. [PMID: 34944392 PMCID: PMC8698585 DOI: 10.3390/biom11121749] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 12/24/2022] Open
Abstract
Programmed cell death 1 ligand (PD-L1) blockade has been used therapeutically in the treatment of ovarian cancer, and potential combination treatment approaches are under investigation to improve the treatment response rate. The increased dependence on glutamine is widely observed in various type of tumors, including ovarian cancer. Kidney-type glutaminase (GLS), as one of the isotypes of glutaminase, is found to promote tumorigenesis. Here, we have demonstrated that the combined treatment with GLS inhibitor 968 and PD-L1 blockade enhances the immune response against ovarian cancer. Survival analysis using the Kaplan–Meier plotter dataset from ovarian cancer patients revealed that the expression level of GLS predicts poor survival and correlates with the immunosuppressive microenvironment of ovarian cancer. 968 inhibits the proliferation of ovarian cancer cells and enhances granzyme B secretion by CD8+ T cells as detected by XTT assay and flow cytometry, respectively. Furthermore, 968 enhances the apoptosis-inducing ability of CD8+ T cells toward cancer cells and improves the treatment effect of anti-PD-L1 in treating ovarian cancer as assessed by Annexin V apoptosis assay. In vivo studies demonstrated the prolonged overall survival upon combined treatment of 968 with anti-PD-L1 accompanied by increased granzyme B secretion by CD4+ and CD8+ T cells isolated from ovarian tumor xenografts. Additionally, 968 increases the infiltration of CD3+ T cells into tumors, possibly through enhancing the secretion of CXCL10 and CXCL11 by tumor cells. In conclusion, our findings provide a novel insight into ovarian cancer cells influence the immune system in the tumor microenvironment and highlight the potential clinical implication of combination of immune checkpoints with GLS inhibitor 968 in treating ovarian cancer.
Collapse
|
41
|
Liao C, Wang A, Ma Y, Liu H. Long non-coding RNA FOXP4-AS1 is a prognostic biomarker and associated with immune infiltrates in ovarian serous cystadenocarcinoma. Medicine (Baltimore) 2021; 100:e27473. [PMID: 34622876 PMCID: PMC8500601 DOI: 10.1097/md.0000000000027473] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 09/21/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND FOXP4-AS1 expression participates in multiple signal pathways and has been previously reported in colorectal cancer, cervical cancer, and other cancer cells. However, its role on prognosis and immune infiltrates in ovarian serous cystadenocarcinoma (OVs) remains unclear. The purpose of our study was to investigate the expression of FOXP4-AS1 in OVs and its association with immune infiltrates, and determined its prognostic roles in OVs. METHODS Using The Cancer Genome Atlas (TCGA) database, we retrieved FOXP4-AS1 expression and clinical information for 376 patients with OVs. Wilcoxon rank sum test was used to compare the expression of FOXP4-AS1 in OVs and normal ovarian tissue. Logistic regression was used to analyze the relationship between clinicopathologic features and FOXP4-AS1. Gene Set Enrichment Analysis (GSEA), and single sample Gene Set Enrichment Analysis (ssGSEA) was conducted to investigate the enrich pathways and functions and quantify the extent of immune cells infiltration for FOXP4-AS1. Kaplan-Meier method was used to generate survival curves, and Cox regression was used to analyze the relationship between FOXP4-AS1 and survival rate. RESULTS High FOXP4-AS1 expression was significantly correlated with tumor FIGO stage (P = .026). Multivariate survival analysis showed that FOXP4-AS1was an independent prognostic marker for overall survival (OS; hazard ratio [HR]: 0.638; 95% confidence interval [CI]:0.467-0.871; P = .001) and disease-specific survival (DSS; HR: 0.649; CI: 0.476-0.885; P = .006). GSEA showed that High FOXP4-AS1 expression may active programmed cell death 1 (PD-1) signaling, the cytotoxic T lymphocyte-associated antigen-4 (CTLA4) pathway, the B cell receptor signaling pathway, apoptosis, fibroblast growth factor receptor (FGFR) signaling, and the Janus-activated kinase signal transducers and activators of transcription (JAK-STAT) signaling pathway. FOXP4-AS1 expression was negatively correlated with markers of immune cells, including aDC, cytotoxic cells and neutrophils. CONCLUSION High FOXP4-AS1 expression has the potential to be a prognostic molecular marker of favorable survival in OVs.
Collapse
Affiliation(s)
- Cheng Liao
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P. R. China
| | - Ao Wang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P. R. China
| | - Yushan Ma
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P. R. China
| | - Hui Liu
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P. R. China
| |
Collapse
|
42
|
Lecuelle J, Boidot R, Mananet H, Derangère V, Albuisson J, Goussot V, Arnould L, Tharin Z, Ray Coquard I, Ghiringhelli F, Truntzer C, Fumet JD. TCR Clonality and Genomic Instability Signatures as Prognostic Biomarkers in High Grade Serous Ovarian Cancer. Cancers (Basel) 2021; 13:4394. [PMID: 34503204 PMCID: PMC8430641 DOI: 10.3390/cancers13174394] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/20/2021] [Accepted: 08/27/2021] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Immune infiltration is a prognostic factor in high-grade serous ovarian carcinoma (HGSC) but immunotherapy efficacy is disappointing. Genomic instability is now used to guide the therapeutic value of PARP inhibitors. We aimed to investigate exome-derived parameters to assess the tumor microenvironment according to genomic instability profile. METHODS We used the HGSC TCGA (the cancer genome atlas) dataset with genomic characteristics, including homologous recombination deficiency (HRD), copy number variant (CNV) signatures, TCR (T cell receptor) clonality and abundance of tissue-infiltrating immune and stromal cell populations. We then investigated the relationship with survival data. RESULTS In 578 HGSC patients, HRD status, CNV signature 7 and TCR clonality were associated with longer survival. The combination of high CNV signature 7 expression and HRD status or high CNV signature 3 expression and high TCR clonality was associated with a trend towards longer survival compared to each variable alone. Combining T cell infiltrate and TCR clonality improved the prognostic value compared to T cells infiltration alone. Prognostic value of TCR clonality was confirmed in an independent cohort. CONCLUSIONS TCR clonality is an emerging prognostic biomarker that improves T cell infiltrate information. Analysis of TCR clonality combined with genomic instability could be an interesting prognostic biomarker.
Collapse
Affiliation(s)
- Julie Lecuelle
- Platform of Transfer in Biological Oncology, Georges François Leclerc Cancer Center—UNICANCER, 1 rue du Professeur Marion, 21000 Dijon, France; (J.L.); (H.M.); (V.D.); (J.A.); (F.G.); (C.T.)
- Unité Mixte de Recherche (UMR) INSERM 1231, 7 Boulevard Jeanne d’Arc, 21000 Dijon, France
| | - Romain Boidot
- Institut de Chimie Moléculaire Université de Bourgogne (ICMUB) UMR CNRS 6302, 21000 Dijon, France;
| | - Hugo Mananet
- Platform of Transfer in Biological Oncology, Georges François Leclerc Cancer Center—UNICANCER, 1 rue du Professeur Marion, 21000 Dijon, France; (J.L.); (H.M.); (V.D.); (J.A.); (F.G.); (C.T.)
| | - Valentin Derangère
- Platform of Transfer in Biological Oncology, Georges François Leclerc Cancer Center—UNICANCER, 1 rue du Professeur Marion, 21000 Dijon, France; (J.L.); (H.M.); (V.D.); (J.A.); (F.G.); (C.T.)
- Department of Biology and Pathology of Tumors, Georges François Leclerc Cancer Center—UNICANCER, 1 rue du Professeur Marion, 21000 Dijon, France; (V.G.); (L.A.)
| | - Juliette Albuisson
- Platform of Transfer in Biological Oncology, Georges François Leclerc Cancer Center—UNICANCER, 1 rue du Professeur Marion, 21000 Dijon, France; (J.L.); (H.M.); (V.D.); (J.A.); (F.G.); (C.T.)
- Department of Biology and Pathology of Tumors, Georges François Leclerc Cancer Center—UNICANCER, 1 rue du Professeur Marion, 21000 Dijon, France; (V.G.); (L.A.)
| | - Vincent Goussot
- Department of Biology and Pathology of Tumors, Georges François Leclerc Cancer Center—UNICANCER, 1 rue du Professeur Marion, 21000 Dijon, France; (V.G.); (L.A.)
| | - Laurent Arnould
- Department of Biology and Pathology of Tumors, Georges François Leclerc Cancer Center—UNICANCER, 1 rue du Professeur Marion, 21000 Dijon, France; (V.G.); (L.A.)
| | - Zoé Tharin
- Department of Medical Oncology, Georges François Leclerc Cancer Center—UNICANCER, 1 rue du Professeur Marion, 21000 Dijon, France;
| | - Isabelle Ray Coquard
- Laboratoire RESHAPE University Claude Bernard Lyon I, Department of Medical Oncology, Léon-Bérard Center, 28 rue Laennec, 69008 Lyon, France;
| | - François Ghiringhelli
- Platform of Transfer in Biological Oncology, Georges François Leclerc Cancer Center—UNICANCER, 1 rue du Professeur Marion, 21000 Dijon, France; (J.L.); (H.M.); (V.D.); (J.A.); (F.G.); (C.T.)
- Unité Mixte de Recherche (UMR) INSERM 1231, 7 Boulevard Jeanne d’Arc, 21000 Dijon, France
- Department of Medical Oncology, Georges François Leclerc Cancer Center—UNICANCER, 1 rue du Professeur Marion, 21000 Dijon, France;
- Maison de l’université Esplanade Erasme, University of Burgundy-Franche Comté, 21000 Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon University Hospital, 14 rue Paul Gaffarel, 21000 Dijon, France
| | - Caroline Truntzer
- Platform of Transfer in Biological Oncology, Georges François Leclerc Cancer Center—UNICANCER, 1 rue du Professeur Marion, 21000 Dijon, France; (J.L.); (H.M.); (V.D.); (J.A.); (F.G.); (C.T.)
- Unité Mixte de Recherche (UMR) INSERM 1231, 7 Boulevard Jeanne d’Arc, 21000 Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon University Hospital, 14 rue Paul Gaffarel, 21000 Dijon, France
| | - Jean-David Fumet
- Platform of Transfer in Biological Oncology, Georges François Leclerc Cancer Center—UNICANCER, 1 rue du Professeur Marion, 21000 Dijon, France; (J.L.); (H.M.); (V.D.); (J.A.); (F.G.); (C.T.)
- Unité Mixte de Recherche (UMR) INSERM 1231, 7 Boulevard Jeanne d’Arc, 21000 Dijon, France
- Department of Medical Oncology, Georges François Leclerc Cancer Center—UNICANCER, 1 rue du Professeur Marion, 21000 Dijon, France;
| |
Collapse
|
43
|
Zhao HB, Zeng YR, Han ZD, Zhuo YJ, Liang YK, Hon CT, Wan S, Wu S, Dahl D, Zhong WD, Wu CL. Novel immune-related signature for risk stratification and prognosis in prostatic adenocarcinoma. Cancer Sci 2021; 112:4365-4376. [PMID: 34252262 PMCID: PMC8486177 DOI: 10.1111/cas.15062] [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: 02/06/2021] [Revised: 06/16/2021] [Accepted: 07/06/2021] [Indexed: 12/14/2022] Open
Abstract
A substantial proportion of prostatic adenocarcinoma (PRAD) patients experience biochemical failure (BCF) after radical prostatectomy (RP). The immune microenvironment plays a vital role in carcinogenesis and the development of PRAD. This study aimed to identify a novel immune-related gene (IRG)-based signature for risk stratification and prognosis of BCF in PRAD. Weighted gene coexpression network analysis was carried out to identify a BCF-related module in a discovery cohort of patients who underwent RP at the Massachusetts General Hospital. The median follow-up time was 70.32 months. Random forest and multivariate stepwise Cox regression analyses were used to identify an IRG-based signature from the specific module. Risk plot analyses, Kaplan-Meier curves, receiver operating characteristic curves, univariate and multivariate Cox regression analyses, stratified analysis, and Harrell's concordance index were used to assess the prognostic value and predictive accuracy of the IRG-based signature in the internal discovery cohort; The Cancer Genome Atlas database was used as a validation cohort. Tumor immune estimation resource database analysis and CIBERSORT algorithm were used to assess the immunophenotype of PRAD. A novel IRG-based signature was identified from the specific module. Five IRGs (BUB1B, NDN, NID1, COL4A6, and FLRT2) were verified as components of the risk signature. The IRG-based signature showed good prognostic value and predictive accuracy in both the discovery and validation cohorts. Infiltrations of various immune cells were significantly different between low-risk and high-risk groups in PRAD. We identified a novel IRG-based signature that could function as an index for assessing tumor immune status and risk stratification in PRAD.
Collapse
Affiliation(s)
- Hai-Bo Zhao
- Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yan-Ru Zeng
- Department of Anesthesiology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.,Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Zhao-Dong Han
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.,Department of Urology, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Yang-Jia Zhuo
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.,Department of Urology, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Ying-Ke Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.,Department of Urology, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Chi Tin Hon
- Macau Institute of Systems Engineering, Macau University of Science and Technology, Macau, China
| | - Song Wan
- Department of Urology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Shulin Wu
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Douglas Dahl
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Wei-De Zhong
- Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.,Department of Urology, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China.,Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Chin-Lee Wu
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
44
|
Marchetti C, D'Indinosante M, Bottoni C, Di Ilio C, Di Berardino S, Costantini B, Minucci A, Vertechy L, Scambia G, Fagotti A. NLR and BRCA mutational status in patients with high grade serous advanced ovarian cancer. Sci Rep 2021; 11:11125. [PMID: 34045513 PMCID: PMC8159985 DOI: 10.1038/s41598-021-90361-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022] Open
Abstract
Laboratory-markers of the systemic inflammatory-response, such as neutrophil/lymphocyte-ratio (NLR) have been studied as prognostic factors in several tumors but in OC-patients their role is still controversial and no data about the possible correlation with the BRCA-status has been ever reported. We consecutively enrolled a series of 397 newly diagnosed high-grade serous-advanced OC-patients. All patients were tested for BRCA-mutational-status and blood-parameters have been collected 48 h before staging-surgery. A significant correlation of NLR with disease distribution (p < 0.005) was found and patients with NLR < 4 underwent primary-debulking-surgery more frequently (p-value 0.001), with a lower surgical-complexity-score (p-value 0.002). Regarding survival-data, patients with NLR < 4 had a significant 7-month increase in mPFS (26 vs 19 months, p = 0.009); focusing on the BRCA-status, among both BRCA-mutated and BRCA-wild type patients, those with lower NLR had a significantly prolonged mPFS compared to patients with NLR > 4 (BRCA-mutated: 35 vs 23 months, p = 0.03; BRCA-wt: 19 vs 16 months, p = 0.05). At multivariate-analysis, independent factors of prolonged PFS were BRCA mutational status, having received complete cytoreduction and NLR < 4. Also, the strongest predictors of longer OS were BRCA-mutational status, having received complete cytoreductive surgery, NLR < 4 and age. NLR is confirmed to be a prognostic marker in OC-patients and it seems unrelated with BRCA-mutational status.
Collapse
Affiliation(s)
- Claudia Marchetti
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marco D'Indinosante
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Catholic University of the Sacred Heart, Rome, Italy
| | - Carolina Bottoni
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Chiara Di Ilio
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Catholic University of the Sacred Heart, Rome, Italy
| | - Stefano Di Berardino
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Catholic University of the Sacred Heart, Rome, Italy
| | - Barbara Costantini
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Angelo Minucci
- Molecular and Genomic Diagnostics Laboratory, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Laura Vertechy
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giovanni Scambia
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy. .,Catholic University of the Sacred Heart, Rome, Italy. .,Department of Women's and Children's Health, Fondazione "Policlinico Universitario A.Gemelli"-IRCCS, Largo Agostino Gemelli 8, 00168, Rome, Italy.
| | - Anna Fagotti
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
45
|
Liu S, Wu M, Wang F. Research Progress in Prognostic Factors and Biomarkers of Ovarian Cancer. J Cancer 2021; 12:3976-3996. [PMID: 34093804 PMCID: PMC8176232 DOI: 10.7150/jca.47695] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer is a serious threat to women's health; its early diagnosis rate is low and prone to metastasis and recurrence. The current conventional treatment for ovarian cancer is a combination of platinum and paclitaxel chemotherapy based on surgery. The recurrence and progression of ovarian cancer with poor prognosis is a major challenge in treatment. With rapid advances in technology, understanding of the molecular pathways involved in ovarian cancer recurrence and progression has increased, biomarker-guided treatment options can greatly improve the prognosis of patients. This review systematically discusses and summarizes existing and new information on prognostic factors and biomarkers of ovarian cancer, which is expected to improve the clinical management of patients and lead to effective personalized treatment.
Collapse
Affiliation(s)
- Shuna Liu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China, 210029
- National Key Clinical Department of Laboratory Medicine, Nanjing, China, 210029
| | - Ming Wu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China, 210029
- National Key Clinical Department of Laboratory Medicine, Nanjing, China, 210029
| | - Fang Wang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China, 210029
- National Key Clinical Department of Laboratory Medicine, Nanjing, China, 210029
| |
Collapse
|
46
|
Zhao X, Zhao B, Zhao Y, Zhang Y, Qian M. Protective effect of anisodamine on bleomycin-induced acute lung injury in immature rats via modulating oxidative stress, inflammation, and cell apoptosis by inhibiting the JAK2/STAT3 pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:859. [PMID: 34164493 PMCID: PMC8184412 DOI: 10.21037/atm-21-1750] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Pediatric acute lung injury (ALI) is one of the most common causes of infant mortality. Although lung-protective strategies have developed in recent years, no ALI treatment is currently available. Anisodamine (Ani) is a common drug used to treat gastrointestinal smooth muscle spasm. The protective effects of Ani against acute kidney injury and myocardial injury have been reported. However, the efficacy of Ani on bleomycin (BLM)-induced ALI has not been examined previously. In the present study, we aimed to examine the effects of Ani on bleomycin (BLM)-induced ALI on immature rats. Methods The ALI rat model was established by intratracheally administration of BLM. Ani treatment was performed by an intravenous injection at different concentrations. The lung function of each rat was measured, and then lung tissue structures, apoptosis, and collagen deposition were observed by hematoxylin-eosin staining, terminal deoxynucleotidyl transferase-mediated digoxigenin-dUTP nick-end labeling assay, and Masson’s staining, respectively. Enzyme-linked immunosorbent assay was used to detect the levels of inflammatory cytokines. The expression of apoptosis-related proteins and fibrosis-related markers was determined by reverse transcription-polymerase chain reaction and/or Western blot analysis. Finally, the expression levels of Janus tyrosine kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3) were determined. Results Our findings indicated that lung function was remarkably decreased in BLM-induced rats, which could be reversed by Ani. Ani treatment increased the levels of antioxidant enzymes, decreased the apoptotic rate and apoptosis-related proteins, and downregulated the expression of fibrosis-related markers. Additionally, Ani treatment also attenuated inflammatory response and suppressed the activation of the JAK2/STAT3 pathway. Conclusions Our results demonstrated that Ani had potent activity against BLM-induced ALI in immature rats through inhibiting the JAK2/STAT3 signaling pathway. Our findings provide supporting evidence to further investigate the therapeutic effect of Ani against ALI in children.
Collapse
Affiliation(s)
- Xiaoqi Zhao
- Department of Neonatology, the Second Hospital of Jilin University, Changchun, China
| | - Bin Zhao
- Department of Neurosurgery, the Second Hospital of Jilin University, Changchun, China
| | - Yinghao Zhao
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun, China
| | - Yunfeng Zhang
- Department of Neonatology, the Second Hospital of Jilin University, Changchun, China
| | - Min Qian
- Department of Neonatology, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
47
|
Batman S, Matsuo K, Mhawech-Fauceglia P, Munro E, Weisenberger M, Allen A, Joshi S, Machida H, Matsuzaki S, Bozanovic T, Pejovic T. Intersection of DNA Repair Pathways and the Immune Landscape Identifies PD-L2 as a Prognostic Marker in Epithelial Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13081972. [PMID: 33923934 PMCID: PMC8073346 DOI: 10.3390/cancers13081972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/12/2021] [Accepted: 04/15/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Here, we examined the interaction between DNA repair proteins and immune biomarkers and their association with survival in 181 cases of epithelial ovarian carcinoma (EOC). We used a panel of 12 antibodies for immunocytochemistry staining of tissue microarray (TMA) consisting of 181 cases. Applying standard statistical methods, we detected that PD-L2 expression was associated with decreased survival in ovarian cancer. This is the first demonstration that increased expression of PD-L2 may serve as a marker for decreased progression-free survival (PFS). Therefore, further investigation into PD-L2 based immunotherapy as a strategy to treat ovarian cancer is warranted. Abstract Background: Targeting DNA repair and immune checkpoint pathways has been the focus of multiple clinical trials. In this study, we explore the association between DNA repair proteins, immune response markers, and clinical outcome in women with EOC. Methods: Immunohistochemical analysis of TMA with 181 EOC samples was used to determine expression levels for DNA repair proteins (PARP, PTEN, p53, H2Ax, FANCD2, and ATM) and immune-markers (CD4, CD8, CD68, PD-L2, PD-L1, and FOXP3). Biomarker expression was correlated to clinical data. Prognostic discriminatory ability was assessed per the combination of biomarkers. Results: Tumor immunity biomarkers correlated with HRD biomarkers. High PD-L2 was significantly associated with high expression of CD8 (r = 0.18), CD68 (r = 0.17), and FOXp3 (r = 0.16) (all, p < 0.05). In a multivariate analysis, PD-L2 (hazard ratio (HR) 1.89), PARP (HR 1.75), and PTEN (HR 1.96) expressions were independently associated with decreased progression-free survival (PFS), whereas PD-L1 (HR 0.49) and CD4 (HR 0.67) were associated with improved PFS (all, p < 0.05). In 15 biomarker combinations, six combinations exhibited a discriminatory ability of >20% for the 4.5-year PFS rate, with four based on PD-L2 (PARP, PTEN, CD4, and PD-L1, 20.5–30.0%). Conclusions: Increased PD-L2 expression is a prognostic marker of decreased survival in EOC. Interaction between tumor DNA repair and microenvironment determines tumor progression and survival.
Collapse
Affiliation(s)
- Samantha Batman
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239-3098, USA; (S.B.); (E.M.); (M.W.); (A.A.); (S.J.)
| | - Koji Matsuo
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA 90033, USA; (K.M.); (P.M.-F.); (S.M.)
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Paulette Mhawech-Fauceglia
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA 90033, USA; (K.M.); (P.M.-F.); (S.M.)
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
- Aurora Diagnostic Center of Excellence in Gynecologic Pathology, Austin, TX 78758, USA
| | - Elizabeth Munro
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239-3098, USA; (S.B.); (E.M.); (M.W.); (A.A.); (S.J.)
| | - Mercedes Weisenberger
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239-3098, USA; (S.B.); (E.M.); (M.W.); (A.A.); (S.J.)
| | - Allison Allen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239-3098, USA; (S.B.); (E.M.); (M.W.); (A.A.); (S.J.)
| | - Sonali Joshi
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239-3098, USA; (S.B.); (E.M.); (M.W.); (A.A.); (S.J.)
| | - Hiroko Machida
- Tokai University School of Medicine, Tokyo 151-8677, Japan;
| | - Shinya Matsuzaki
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA 90033, USA; (K.M.); (P.M.-F.); (S.M.)
| | - Tatjana Bozanovic
- Department of Obstetrics and Gynecology, University of Belgrade, 11000 Belgrade, Serbia;
| | - Tanja Pejovic
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239-3098, USA; (S.B.); (E.M.); (M.W.); (A.A.); (S.J.)
- Correspondence: ; Tel.: +1-503-494-4500
| |
Collapse
|
48
|
Sawada R, Akiyoshi T, Kitagawa Y, Hiyoshi Y, Mukai T, Nagasaki T, Yamaguchi T, Konishi T, Yamamoto N, Ueno M, Fukunaga Y. Systemic Inflammatory Markers Combined with Tumor-Infiltrating Lymphocyte Density for the Improved Prediction of Response to Neoadjuvant Chemoradiotherapy in Rectal Cancer. Ann Surg Oncol 2021; 28:6189-6198. [PMID: 33876358 DOI: 10.1245/s10434-021-09975-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/24/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Previous studies have reported the utility of systemic inflammatory markers and CD8+ tumor-infiltrating lymphocyte (TIL) separately in predicting response to chemoradiotherapy (CRT) in rectal cancer; however, the efficacy of combining these markers remains unclear. OBJECTIVE This study aimed to elucidate the predictive efficacy of systemic inflammatory markers combined with CD8+ TIL density on response to neoadjuvant CRT in locally advanced rectal cancer. METHODS Ten systemic inflammatory markers and CD8+ TIL density were assessed in 267 patients with rectal cancer using pretreatment clinical data and biopsy samples. Response to CRT was determined using the Dworak tumor regression grade (TRG), with good responders classified as TRG3-4. RESULTS Receiver operating characteristic curve analysis showed high areas under the curve for the lymphocyte-to-C-reactive protein ratio (LCR) and neutrophil × monocyte (N × M) value (0.58 and 0.62, respectively). In the multivariate analysis, LCR, N × M value, and CD8+ TIL density were independently associated with good responders (p = 0.016, 0.005, and 0.002, respectively). Stratified analysis with these three markers showed a positive correlation between TRG3-4 ratio and the number of positive predictive factors (8.2%, 20.0%, 34.2%, and 59.1% in patients with 0, 1, 2, and 3 predictors, respectively). Overall and disease-free survival were significantly worse in patients with zero factors present compared with those with one to three factors present. CONCLUSIONS LCR, N × M value, and CD8+ TIL density are independently associated with response to CRT. Assessing local TIL density along with systemic inflammatory markers may be useful for selecting a multimodal neoadjuvant approach in rectal cancer therapy.
Collapse
Affiliation(s)
- Ryuichiro Sawada
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takashi Akiyoshi
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Yusuke Kitagawa
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yukiharu Hiyoshi
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Toshiki Mukai
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Toshiya Nagasaki
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tomohiro Yamaguchi
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tsuyoshi Konishi
- Department of Colon and Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Noriko Yamamoto
- Department of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masashi Ueno
- Department of Gastroenterological Surgery, Toranomon Hospital, Tokyo, Japan
| | - Yosuke Fukunaga
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
49
|
Noninvasive prediction of residual disease for advanced high-grade serous ovarian carcinoma by MRI-based radiomic-clinical nomogram. Eur Radiol 2021; 31:7855-7864. [PMID: 33864139 DOI: 10.1007/s00330-021-07902-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 02/13/2021] [Accepted: 03/16/2021] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To develop a preoperative MRI-based radiomic-clinical nomogram for prediction of residual disease (RD) in patients with advanced high-grade serous ovarian carcinoma (HGSOC). METHODS In total, 217 patients with advanced HGSOC were enrolled from January 2014 to June 2019 and randomly divided into a training set (n = 160) and a validation set (n = 57). Finally, 841 radiomic features were extracted from each tumor on T2-weighted imaging (T2WI) and contrast-enhanced T1-weighted imaging (CE-T1WI) sequence, respectively. We used two fusion methods, the maximal volume of interest (MV) and the maximal feature value (MF), to fuse the radiomic features of bilateral tumors, so that patients with bilateral tumors have the same kind of radiomic features as patients with unilateral tumors. The radiomic signatures were constructed by using mRMR method and LASSO classifier. Multivariable logistic regression analysis was used to develop a radiomic-clinical nomogram incorporating radiomic signature and conventional clinico-radiological features. The performance of the nomogram was evaluated on the validation set. RESULTS In total, 342 tumors from 217 patients were analyzed in this study. The MF-based radiomic signature showed significantly better prediction performance than the MV-based radiomic signature (AUC = 0.744 vs. 0.650, p = 0.047). By incorporating clinico-radiological features and MF-based radiomic signature, radiomic-clinical nomogram showed favorable prediction ability with an AUC of 0.803 in the validation set, which was significantly higher than that of clinico-radiological signature and MF-based radiomic signature (AUC = 0.623, 0.744, respectively). CONCLUSIONS The proposed MRI-based radiomic-clinical nomogram provides a promising way to noninvasively predict the RD status. KEY POINTS • MRI-based radiomic-clinical nomogram is feasible to noninvasively predict residual disease in patients with advanced HGSOC. • The radiomic signature based on MF showed significantly better prediction performance than that based on MV. • The radiomic-clinical nomogram showed a favorable prediction ability with an AUC of 0.803.
Collapse
|
50
|
Jain S, Annett SL, Morgan MP, Robson T. The Cancer Stem Cell Niche in Ovarian Cancer and Its Impact on Immune Surveillance. Int J Mol Sci 2021; 22:4091. [PMID: 33920983 PMCID: PMC8071330 DOI: 10.3390/ijms22084091] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/11/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer is an aggressive gynaecological cancer with extremely poor prognosis, due to late diagnosis as well as the development of chemoresistance after first-line therapy. Research advances have found stem-like cells present in ovarian tumours, which exist in a dynamic niche and persist through therapy. The stem cell niche interacts extensively with the immune and non-immune components of the tumour microenvironment. Significant pathways associated with the cancer stem cell niche have been identified which interfere with the immune component of the tumour microenvironment, leading to immune surveillance evasion, dysfunction and suppression. This review aims to summarise current evidence-based knowledge on the cancer stem cell niche within the ovarian cancer tumour microenvironment and its effect on immune surveillance. Furthermore, the review seeks to understand the clinical consequences of this dynamic interaction by highlighting current therapies which target these processes.
Collapse
Affiliation(s)
| | | | | | - Tracy Robson
- School of Pharmacy and Biomolecular Science, RCSI University of Medicine and Health Sciences, 123 St Stephen’s Green, D02 YN77 Dublin, Ireland; (S.J.); (S.L.A.); (M.P.M.)
| |
Collapse
|