1
|
Zeng P, Lu L, Zhang H, Li Y, Tan S, Yu T, Zhou H. Therapeutic targets for endometriosis: Genome-wide Mendelian randomization and colocalization analyses. Gene 2024; 893:147970. [PMID: 37931855 DOI: 10.1016/j.gene.2023.147970] [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: 08/29/2023] [Revised: 10/09/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Endometriosis (EM) greatly affects women's reproductive health, identifying new drug targets for EM is urgently needed. This study utilizes comprehensive genome-wide Mendelian randomization (MR) and colocalization analyses, using genomic data, to identify potential therapeutic approaches for EM. METHODS Genome-wide cis-expression quantitative trait loci (cis-eQTL) data were obtained from GTEx V8, which included 838 participants across 49 tissues or cells, and the eQTLGen consortium, which included 31,684 participants. Genome-wide association analysis (GWAS) data for EM were sourced from the FinnGen study, which consisted of 8,288 cases and 68,969 controls, as well as the UK Biobank study, which included 1,496 cases and 359,698 controls. This study utilized MR analysis to assess the correlation between genes and the risk of EM. Subsequently, colocalization analysis was conducted to investigate potential shared causal variants between the identified genes and EM. RESULTS After conducting MR and colocalization analyses, we identified a total of 13 genes that showed significant evidence of colocalization. These genes are considered promising therapeutic candidates for treating EM. Among them, inner membrane mitochondrial protein (IMMT), src kinase associated phosphoprotein 1 (SKAP1), lysine methyltransferase 5A (KMT5A), KLF transcription factor 12 (KLF12), GRB10 interacting GYF protein 1 (GIGYF1), Wnt family member 7A (WNT7A), Sad1 and UNC84 domain containing 1 (SUN1), and poly (ADP-ribose) polymerase family member 3 (PARP3) were found to have positive associations with the risk of EM. On the other hand, progestin and adipoQ receptor family member 8 (PAQR8), adaptor related protein complex 3 subunit mu 1 (AP3M1), surfeit 6 (SURF6), TUB bipartite transcription factor (TUB), and DNA polymerase delta interacting protein 2 (POLDIP2) were found to have inverse relationships with the risk of EM. CONCLUSIONS Through genome-wide MR studies, a comprehensive set of genes associated with EM has been identified. Among them, IMMT, PAQR8, SKAP1, KMT5A, AP3M1, SURF6, KLF12, GIGYF1, TUB, WNT7A, SUN1, POLDIP2, and PARP3 show potential as therapeutic targets for EM treatment. Nonetheless, it is crucial to conduct further rigorous investigations to validate these prospects.
Collapse
Affiliation(s)
- Pengfei Zeng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Liyue Lu
- School of Shuguang Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hanxiao Zhang
- Faculty of Medicine, Université Paris-Saclay, Villejuif, France
| | - Yanting Li
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shufa Tan
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xi'an, Sichuan, China
| | - Tong Yu
- Department of Gynecology, Guangan Hospital of Traditional Chinese Medicine, Guangan, Sichuan, China.
| | - Hang Zhou
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Xagara A, Roumeliotou A, Kokkalis A, Tsapakidis K, Papakonstantinou D, Papadopoulos V, Samaras I, Chantzara E, Kallergi G, Kotsakis A. ES-SCLC Patients with PD-L1 + CTCs and High Percentages of CD8 +PD-1 +T Cells in Circulation Benefit from Front-Line Immunotherapy Treatment. Biomedicines 2024; 12:146. [PMID: 38255251 PMCID: PMC10813758 DOI: 10.3390/biomedicines12010146] [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: 12/06/2023] [Revised: 12/29/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
SCLC is an aggressive cancer type with high metastatic potential and bad prognosis. CTCs are a valuable source of tumor cells in blood circulation and are among the major contributors to metastasis. In this study we evaluated the number of CTCs that express PD-L1 in treatment-naïve ES-SCLC patients receiving ICI in a front-line setting. Moreover, we explored the percentages of different immune T-cell subsets in circulation to assess their potential role in predicting responses. A total of 43 patients were enrolled-6 of them with LS-SCLC, and 37 with ES-SCLC disease. In addition, PBMCs from 10 healthy donors were used as a control group. Different T-cell subtypes were examined through multicolor FACS analysis and patients' CTCs were detected using immunofluorescence staining. SCLC patients had higher percentages of PD-1-expressing CD3+CD4+ and CD3+CD8+ T-cells, as well as elevated PD-1 protein expression compared to healthy individuals. Additionally, in ES-SCLC patients, a positive correlation between CD3+CD8+PD-1+ T-cells and PD-L1+ CTCs was detected. Importantly, patients harboring higher numbers of CD3+CD8+PD-1+ T-cells together with PD-L1+CTCs had a survival advantage when receiving front-line immunotherapy. Thus, this study proposes, for first time possible, immune cell-CTCs interaction, as well as a potential novel clinical biomarker for ICI responses in ES-SCLC patients.
Collapse
Affiliation(s)
- Anastasia Xagara
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.); (K.T.); (V.P.); (I.S.); (E.C.)
| | - Argyro Roumeliotou
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece; (A.R.); (D.P.); (G.K.)
| | - Alexandros Kokkalis
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.); (K.T.); (V.P.); (I.S.); (E.C.)
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Konstantinos Tsapakidis
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.); (K.T.); (V.P.); (I.S.); (E.C.)
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Dimitris Papakonstantinou
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece; (A.R.); (D.P.); (G.K.)
| | - Vassilis Papadopoulos
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.); (K.T.); (V.P.); (I.S.); (E.C.)
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Ioannis Samaras
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.); (K.T.); (V.P.); (I.S.); (E.C.)
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Evagelia Chantzara
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.); (K.T.); (V.P.); (I.S.); (E.C.)
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Galatea Kallergi
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece; (A.R.); (D.P.); (G.K.)
| | - Athanasios Kotsakis
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.); (K.T.); (V.P.); (I.S.); (E.C.)
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| |
Collapse
|
3
|
Acheampong E, Allsopp RC, Page K, Wadsley MK, Beasley AB, Coombes RC, Shaw JA, Gray ES. Meta-Analysis of Circulating Tumor Cell PD-L1 Expression and the Association with Clinical Outcomes in Non-Small Cell Lung Cancer. Clin Chem 2024; 70:234-249. [PMID: 38175603 DOI: 10.1093/clinchem/hvad187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/23/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Programmed death ligand-1 (PD-L1) expression on circulating tumor cells (CTCs) has been suggested to provide prognostic information in non-small cell lung cancer (NSCLC), but consensus relative to treatment outcomes is lacking. We conducted the first comprehensive meta-analysis exploring its potential as a prognostic and predictive marker, and assessed the concordance between PD-L1 + CTCs and paired tumor tissue in NSCLC patients. METHOD A comprehensive search was applied to PubMed and EMBASE to identify 26 studies that evaluated PD-L1 + CTCs and their association with survival outcomes in 1236 NSCLC patients. RESULTS The meta-analysis estimated a mean PD-L1 + CTCs detection rate of 61% (95% CI, 49-72). Subgroup analysis based on treatment showed that PD-L1 + CTCs was not significantly associated with better overall survival (OS) in NSCLC patients treated with immune checkpoint inhibitors (ICIs) (Hazard Ratio (HR) = 0.96, 95% CI, 0.35-2.65, P = 0.944), but was predictive of worse OS in those treated with other therapies (HR = 2.11, 95% CI, 1.32-3.36, P = 0.002). Similarly, PD-L1 + CTCs was not significantly associated with superior progressing free survival (PFS) in NSCLCs treated with ICIs (HR = 0.67, 95% CI, 0.41-1.09, P = 0.121), but was significantly associated with shorter PFS in patients treated with other therapies (HR = 1.91, 95% CI, 1.24-2.94, P = 0.001). The overall estimate for the concordance between PD-L1 expression on CTCs and tumor cells was 63% (95% CI, 44-80). CONCLUSION The average detection rate of PD-L1 + CTCs was comparable to the rate of PD-L1 expression in NSCLC tumors. There was a trend towards better PFS in ICI-treated NSCLC patients with PD-L1 + CTCs. Larger longitudinal studies on the association of PD-L1 + CTCs with clinical outcomes in NSCLC patients treated with ICIs are warranted.
Collapse
Affiliation(s)
- Emmanuel Acheampong
- Leicester Cancer Research Centre, Department of Genetic and Genome Biology, University of Leicester, Leicester, United Kingdom
- Institute of Precision Health, University of Leicester, Leicester, United Kingdom
| | - Rebecca C Allsopp
- Leicester Cancer Research Centre, Department of Genetic and Genome Biology, University of Leicester, Leicester, United Kingdom
- Institute of Precision Health, University of Leicester, Leicester, United Kingdom
| | - Karen Page
- Leicester Cancer Research Centre, Department of Genetic and Genome Biology, University of Leicester, Leicester, United Kingdom
- Institute of Precision Health, University of Leicester, Leicester, United Kingdom
| | - Marc K Wadsley
- Leicester Cancer Research Centre, Department of Genetic and Genome Biology, University of Leicester, Leicester, United Kingdom
- Institute of Precision Health, University of Leicester, Leicester, United Kingdom
| | - Aaron B Beasley
- School of Medical and Health Sciences, Edith Cowan University, Perth, Joondalup, WA, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia
| | - R Charles Coombes
- Department of Surgery and Cancer, Imperial College London, Hammersmith, Hospital Campus, London, United Kingdom
| | - Jacqui A Shaw
- Leicester Cancer Research Centre, Department of Genetic and Genome Biology, University of Leicester, Leicester, United Kingdom
- Institute of Precision Health, University of Leicester, Leicester, United Kingdom
| | - Elin S Gray
- School of Medical and Health Sciences, Edith Cowan University, Perth, Joondalup, WA, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia
| |
Collapse
|
4
|
Stoecklein NH, Oles J, Franken A, Neubauer H, Terstappen LWMM, Neves RPL. Clinical application of circulating tumor cells. MED GENET-BERLIN 2023; 35:237-250. [PMID: 38835741 PMCID: PMC11110132 DOI: 10.1515/medgen-2023-2056] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
This narrative review aims to provide a comprehensive overview of the current state of circulating tumor cell (CTC) analysis and its clinical significance in patients with epithelial cancers. The review explores the advancements in CTC detection methods, their clinical applications, and the challenges that lie ahead. By examining the important research findings in this field, this review offers the reader a solid foundation to understand the evolving landscape of CTC analysis and its potential implications for clinical practice. The comprehensive analysis of CTCs provides valuable insights into tumor biology, treatment response, minimal residual disease detection, and prognostic evaluation. Furthermore, the review highlights the potential of CTCs as a non-invasive biomarker for personalized medicine and the monitoring of treatment efficacy. Despite the progress made in CTC research, several challenges such as standardization, validation, and integration into routine clinical practice remain. The review concludes by discussing future directions and the potential impact of CTC analysis on improving patient outcomes and guiding therapeutic decision-making in epithelial cancers.
Collapse
Affiliation(s)
- Nikolas H Stoecklein
- Heinrich-Heine University Düsseldorf General, Visceral and Pediatric Surgery University Hospital and Medical Faculty Düsseldorf Deutschland
| | - Julia Oles
- Heinrich-Heine University Düsseldorf General, Visceral and Pediatric Surgery University Hospital and Medical Faculty Düsseldorf Deutschland
| | - Andre Franken
- University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf Department of Obstetrics and Gynecology Düsseldorf Deutschland
| | - Hans Neubauer
- University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf Department of Obstetrics and Gynecology Düsseldorf Deutschland
| | - Leon W M M Terstappen
- Heinrich-Heine University Düsseldorf General, Visceral and Pediatric Surgery University Hospital and Medical Faculty Düsseldorf Deutschland
| | - Rui P L Neves
- Heinrich-Heine University Düsseldorf General, Visceral and Pediatric Surgery University Hospital and Medical Faculty Düsseldorf Deutschland
| |
Collapse
|
5
|
Hu B, Chen D, Li Y, Yu S, Kuang L, Ma X, Yang Q, He K, Zhao Y, Wang G, Guo M. Expression of TXLNA in brain gliomas and its clinical significance: a bioinformatics analysis. Chin Neurosurg J 2023; 9:27. [PMID: 37752559 PMCID: PMC10521531 DOI: 10.1186/s41016-023-00341-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 08/29/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND To analyze the expression of TXLNA in brain gliomas and its clinical significance. METHODS Gene Expression Profiling Interactive Analysis(GEPIA)and Chinese Glioma Genome Atlas(CGGA)databases were retrieved as the methods. To assess the disparity between TXLNA expression in glioma and normal brain tissue. The Kaplan-Meier survival curve was employed to preliminarily evaluate the survival curves of the high and low expression groups, this was done for investigate the correlation between TXLNA expression level and the survival and prognosis of glioma. A Cox proportional regression risk model of multivariate nature was employed to evaluate the elements impacting the survival and prognosis of glioma. Gene pool enrichment analysis(GSEA)was used to investigate the related function of TXLNA in glioma. A Pearson correlation test and co-expression analysis were employed to identify the genes most associated with TXLNA expression. RESULT The enrichment analysis results were observably enriched in signal pathways for instance the cell cycle and completion and coordination cascade pathways, and it is evident that high expression of TXLNA in gliomas is related to a poor survival and a bad patient prognosis, thus making it an independent prognostic factor for gliomas. Genes such as STK40 and R1MS1 are significantly correlated with TXLNA, playing a synergistic or antagonistic role. CONCLUSIONS The prognosis of GBM patients is strongly linked to the high expression of TXLNA, which may be a viable therapeutic target for curbing cancer progression and creating new immunotherapies for GBM.
Collapse
Affiliation(s)
- Bowen Hu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, Harbin, 150086, Heilongjiang Province, China
| | - Desheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, Harbin, 150086, Heilongjiang Province, China
| | - Yang Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, Harbin, 150086, Heilongjiang Province, China
| | - Shan Yu
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, Harbin, 150086, Heilongjiang Province, China
| | - Liangwen Kuang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, Harbin, 150086, Heilongjiang Province, China
| | - Xinqi Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, Harbin, 150086, Heilongjiang Province, China
| | - Qingsong Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, Harbin, 150086, Heilongjiang Province, China
| | - Ke He
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, Harbin, 150086, Heilongjiang Province, China
| | - Yan Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, Harbin, 150086, Heilongjiang Province, China
| | - Guangzhi Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, Harbin, 150086, Heilongjiang Province, China.
| | - Mian Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, Harbin, 150086, Heilongjiang Province, China.
| |
Collapse
|
6
|
Vardas V, Ju JA, Christopoulou A, Xagara A, Georgoulias V, Kotsakis A, Alix-Panabières C, Martin SS, Kallergi G. Functional Analysis of Viable Circulating Tumor Cells from Triple-Negative Breast Cancer Patients Using TetherChip Technology. Cells 2023; 12:1940. [PMID: 37566019 PMCID: PMC10416943 DOI: 10.3390/cells12151940] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
Metastasis, rather than the growth of the primary tumor, accounts for approximately 90% of breast cancer patient deaths. Microtentacles (McTNs) formation represents an important mechanism of metastasis. Triple-negative breast cancer (TNBC) is the most aggressive subtype with limited targeted therapies. The present study aimed to isolate viable circulating tumor cells (CTCs) and functionally analyze them in response to drug treatment. CTCs from 20 TNBC patients were isolated and maintained in culture for 5 days. Biomarker expression was identified by immunofluorescence staining and VyCap analysis. Vinorelbine-induced apoptosis was evaluated based on the detection of M30-positive cells. Our findings revealed that the CTC absolute number significantly increased using TetherChips analysis compared to the number of CTCs in patients' cytospins (p = 0.006) providing enough tumor cells for drug evaluation. Vinorelbine treatment (1 h) on live CTCs led to a significant induction of apoptosis (p = 0.010). It also caused a significant reduction in Detyrosinated α-tubulin (GLU), programmed death ligand (PD-L1)-expressing CTCs (p < 0.001), and disruption of McTNs. In conclusion, this pilot study offers a useful protocol using TetherChip technology for functional analysis and evaluation of drug efficacy in live CTCs, providing important information for targeting metastatic dissemination at a patient-individualized level.
Collapse
Affiliation(s)
- Vasileios Vardas
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece;
| | - Julia A. Ju
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.A.J.); (S.S.M.)
| | | | - Anastasia Xagara
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.)
| | | | - Athanasios Kotsakis
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece; (A.X.); (A.K.)
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Center of Montpellier, 34295 Montpellier, France;
- CREEC/CANECEV, MIVEGEC (CREES), Université de Montpellier, CNRS, IRD, 34090 Montpellier, France
- European Liquid Biopsy Society (ELBS), 20246 Hamburg, Germany
| | - Stuart S. Martin
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.A.J.); (S.S.M.)
| | - Galatea Kallergi
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece;
| |
Collapse
|
7
|
Vardas V, Tolios A, Christopoulou A, Georgoulias V, Xagara A, Koinis F, Kotsakis A, Kallergi G. Immune Checkpoint and EMT-Related Molecules in Circulating Tumor Cells (CTCs) from Triple Negative Breast Cancer Patients and Their Clinical Impact. Cancers (Basel) 2023; 15:1974. [PMID: 37046635 PMCID: PMC10093450 DOI: 10.3390/cancers15071974] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/19/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Triple negative breast cancer (TNBC) is the most aggressive breast cancer subtype. There are few targeted therapies for these patients, leading to an unmet need for new biomarkers. The present study aimed to investigate the expression of PD-L1, CTLA-4, GLU, and VIM in CTCs of TNBC patients. Ninety-five patients were enrolled in this study: sixty-four TNBC and thirty-one luminal. Of these patients, 60 were in the early stage, while 35 had metastatic disease. Protein expression was identified by immunofluorescence staining experiments and VyCAP analysis. All the examined proteins were upregulated in TNBC patients. The expression of the GLU+VIM+CK+ phenotype was higher (50%) in metastatic TNBC compared to early TNBC patients (17%) (p = 0.005). Among all the BC patients, a significant correlation was found between PD-L1+CD45-CK+ and CTLA-4+CD45-CK+ phenotypes (Spearman test, p = 0.024), implying an important role of dual inhibition in BC. Finally, the phenotypes GLU+VIM+CK+ and PD-L1+CD45-CK+ were associated with shorter OS in TNBC patients (OS: log-rank p = 0.048, HR = 2.9, OS: log-rank p < 0.001, HR = 8.7, respectively). Thus, PD-L1, CTLA-4, GLU, and VIM constitute significant biomarkers in TNBC associated with patients' outcome, providing new therapeutic targets for this difficult breast cancer subtype.
Collapse
Affiliation(s)
- Vasileios Vardas
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece
| | - Anastasios Tolios
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece
| | | | | | - Anastasia Xagara
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece
| | - Filippos Koinis
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Athanasios Kotsakis
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Galatea Kallergi
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece
| |
Collapse
|
8
|
Phenotypic Characterization of Circulating Tumor Cells Isolated from Non-Small and Small Cell Lung Cancer Patients. Cancers (Basel) 2022; 15:cancers15010171. [PMID: 36612166 PMCID: PMC9818148 DOI: 10.3390/cancers15010171] [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: 11/25/2022] [Revised: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
In the present study, we evaluated the expression of JUNB and CXCR4 in circulating tumor cells (CTCs) of lung cancer patients and investigated whether these proteins have prognostic clinical relevance. Peripheral blood from 30 patients with non-small-cell lung cancer (NSCLC) was filtered using ISET membranes, and cytospins from 37 patients with small-cell lung cancer (SCLC) were analyzed using confocal and VyCAP microscopy. Both JUNB and CXCR4 were expressed in the vast majority of lung cancer patients. Interestingly, the phenotypic patterns differed between NSCLC and SCLC patients; the (CK+/JUNB+/CXCR4+) phenotype was present in 50% of NSCLC vs. 71% of SCLC patients. Similarly, the (CK+/JUNB+/CXCR4−) was present in 44% vs. 71%, the (CK+/JUNB−/CXCR4+) in 6% vs. 71%, and the (CK+/JUNB−/CXCR4−) phenotype in 38% vs. 84%. In NSCLC, the presence of ≥1 CTCs with the (CK+/JUNB+/CXCR4+) phenotype was associated with worse progression-free survival (PFS) (p = 0.007, HR = 5.21) while ≥2 with poorer overall survival (OS) (p < 0.001, HR = 2.16). In extensive stage SCLC patients, the presence of ≥4 CXCR4-positive CTCs was associated with shorter OS (p = 0.041, HR = 5.01). Consequently, JUNB and CXCR4 were expressed in CTCs from lung cancer patients, and associated with patients’ survival, underlying their key role in tumor progression.
Collapse
|