1
|
Dadlani E, Dash T, Sahoo D. Investigating tumor-associated macrophages and their polarization in colorectal cancer using Boolean implication networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.08.01.551559. [PMID: 37577482 PMCID: PMC10418212 DOI: 10.1101/2023.08.01.551559] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Tumor-associated Macrophages (or TAMs) are amongst the most common cells that play a significant role in the initiation and progression of colorectal cancer (CRC). [Ghosh et al., 2023] have built a Boolean-logic dependent model to propose a set of gene signatures capable of identifying macrophage polarization states. The signature, called the Signature of Macrophage Reactivity and Tolerance (SMaRT), comprises of 338 human genes (equivalently, 298 mouse genes). The SMaRT signature was constructed using datasets that were not specialized towards any particular disease. To specifically investigate macrophage polarization in CRC, in this paper, we (a) perform a comprehensive analysis of the SMaRT signature on single-cell human and mouse colorectal cancer RNA-seq datasets and (b) adopt transfer learning to construct a "refined" SMaRT signature that specifically characterizes TAM polarization in the CRC tumor microenvironment. Towards validation of our refined gene signature, we use: (a) 5 RNA-seq datasets derived from single-cell human datasets; and (b) 5 large-cohort microarray datasets from humans. Furthermore, we propose the translational potential of our refined gene signature while investigating microsatellite stability and CpG island methylator phenotype (CIMP) in colorectal cancer. Overall, our refined gene signature and its extensive validation provide a path for its adoption in clinical practice in diagnosing colorectal cancer and associated attributes. Availability and Implementation The data, codes, and software packages used in our research are linked and shared publicly at https://github.com/tirtharajdash/TAMs-CRC .
Collapse
|
2
|
Etzi F, Griñán-Lisón C, Fenu G, González-Titos A, Pisano A, Farace C, Sabalic A, Picon-Ruiz M, Marchal JA, Madeddu R. The Role of miR-486-5p on CSCs Phenotypes in Colorectal Cancer. Cancers (Basel) 2024; 16:4237. [PMID: 39766136 PMCID: PMC11674241 DOI: 10.3390/cancers16244237] [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: 11/12/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third diagnosed cancer worldwide. Forty-four percent of metastatic colorectal cancer patients were diagnosed at an early stage. Despite curative resection, approximately 40% of patients will develop metastases within a few years. Previous studies indicate the presence of cancer stem cells (CSCs) and their contribution to CRC progression and metastasis. miRNAs deregulation plays a role in CSCs formation and in tumor development. In light of previous studies, we investigated the role of miR-486-5p to understand its role in CSC better. METHODS The expression of miR-486-5p was assessed in adherent cells and spheres generated from two CRC cell lines to observe the difference in expression in CSC-enriched spheroids. Afterward, we overexpressed and underexpressed this miRNA in adherent and sphere cultures through the transfection of a miR-486-5p mimic and a mimic inhibitor. RESULTS The results demonstrated that miR-486-5p exhibited a notable downregulation in CSC models, and its overexpression led to a significant decrease in colony size. CONCLUSIONS In this study, we confirmed that miR-486-5p plays an oncosuppressive role in CRC, thereby advancing our understanding of the role of this microRNA in the CSC phenotype.
Collapse
Affiliation(s)
- Federica Etzi
- Department of Biomedical Science, University of Sassari, 07100 Sassari, Italy or (F.E.); (G.F.); (C.F.); (A.S.); (R.M.)
| | - Carmen Griñán-Lisón
- Department of Biochemistry and Molecular Biology 2, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012 Granada, Spain; (A.G.-T.); (M.P.-R.); (J.A.M.)
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18100 Granada, Spain
| | - Grazia Fenu
- Department of Biomedical Science, University of Sassari, 07100 Sassari, Italy or (F.E.); (G.F.); (C.F.); (A.S.); (R.M.)
| | - Aitor González-Titos
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012 Granada, Spain; (A.G.-T.); (M.P.-R.); (J.A.M.)
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - Andrea Pisano
- Department of Biomedical Science, University of Sassari, 07100 Sassari, Italy or (F.E.); (G.F.); (C.F.); (A.S.); (R.M.)
| | - Cristiano Farace
- Department of Biomedical Science, University of Sassari, 07100 Sassari, Italy or (F.E.); (G.F.); (C.F.); (A.S.); (R.M.)
| | - Angela Sabalic
- Department of Biomedical Science, University of Sassari, 07100 Sassari, Italy or (F.E.); (G.F.); (C.F.); (A.S.); (R.M.)
| | - Manuel Picon-Ruiz
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012 Granada, Spain; (A.G.-T.); (M.P.-R.); (J.A.M.)
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18100 Granada, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Juan Antonio Marchal
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012 Granada, Spain; (A.G.-T.); (M.P.-R.); (J.A.M.)
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18100 Granada, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Roberto Madeddu
- Department of Biomedical Science, University of Sassari, 07100 Sassari, Italy or (F.E.); (G.F.); (C.F.); (A.S.); (R.M.)
- National Institute of Biostructures and Biosystems, 00136 Rome, Italy
| |
Collapse
|
3
|
Zhang Z, Wang M, Dai R, Wang Z, Lei L, Zhao X, Han K, Shi C, Guo Q. GraphCVAE: Uncovering cell heterogeneity and therapeutic target discovery through residual and contrastive learning. Life Sci 2024; 359:123208. [PMID: 39488267 DOI: 10.1016/j.lfs.2024.123208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/03/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024]
Abstract
Advancements in Spatial Transcriptomics (ST) technologies in recent years have transformed the analysis of tissue structure and function within spatial contexts. However, accurately identifying spatial domains remains challenging due to data sparsity and noise. Traditional clustering methods often fail to capture spatial dependencies, while spatial clustering methods struggle with batch effects and data integration. We introduce GraphCVAE, a model designed to enhance spatial domain identification by integrating spatial and morphological information, correcting batch effects, and managing heterogeneous data. GraphCVAE employs a multi-layer Graph Convolutional Network (GCN) and a variational autoencoder to improve the representation and integration of spatial information. Through contrastive learning, the model captures subtle differences between cell types and states. Extensive testing on various ST datasets demonstrates GraphCVAE's robustness and biological contributions. In the dorsolateral prefrontal cortex (DLPFC) dataset, it accurately delineates cortical layer boundaries. In glioblastoma, GraphCVAE reveals critical therapeutic targets such as TF and NFIB. In colorectal cancer, it explores the role of the extracellular matrix in colorectal cancer. The model's performance metrics consistently surpass existing methods, validating its effectiveness. GraphCVAE's advanced visualization capabilities further highlight its precision in resolving spatial structures, making it a powerful tool for spatial transcriptomics analysis and offering new insights into disease studies.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Mengqiu Wang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Ruoyan Dai
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Zhenghui Wang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Lixin Lei
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Xudong Zhao
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Kaitai Han
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Chaojing Shi
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Qianjin Guo
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China.
| |
Collapse
|
4
|
Wang Q, Greene MI. Survivin as a Therapeutic Target for the Treatment of Human Cancer. Cancers (Basel) 2024; 16:1705. [PMID: 38730657 PMCID: PMC11083197 DOI: 10.3390/cancers16091705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Survivin was initially identified as a member of the inhibitor apoptosis (IAP) protein family and has been shown to play a critical role in the regulation of apoptosis. More recent studies showed that survivin is a component of the chromosome passenger complex and acts as an essential mediator of mitotic progression. Other potential functions of survivin, such as mitochondrial function and autophagy, have also been proposed. Survivin has emerged as an attractive target for cancer therapy because its overexpression has been found in most human cancers and is frequently associated with chemotherapy resistance, recurrence, and poor survival rates in cancer patients. In this review, we discuss our current understanding of how survivin mediates various aspects of malignant transformation and drug resistance, as well as the efforts that have been made to develop therapeutics targeting survivin for the treatment of cancer.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Mark I. Greene
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Jiang X, Wang S, Liang Q, Liu Y, Liu L. Unraveling the multifaceted role of EpCAM in colorectal cancer: an integrated review of its function and interplay with non-coding RNAs. Med Oncol 2023; 41:35. [PMID: 38151631 DOI: 10.1007/s12032-023-02273-6] [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: 09/12/2023] [Accepted: 11/27/2023] [Indexed: 12/29/2023]
Abstract
The epithelial cell adhesion molecule (EpCAM) is a critical glycoprotein involved in cell cycle progression, proliferation, differentiation, migration, and immune evasion. Its role as a target for bispecific antibodies has shown promise in annihilating cancer cells. EpCAM's potential as a biomarker for tumor-initiating cells, characterized by self-renewal and tumorigenic capabilities, underscores its value in early cancer detection, immunotherapy, and targeted drug delivery. While EpCAM monotherapies have been met with limited success, bispecific antibodies targeting both EpCAM and other proteins have exhibited encouraging results in colorectal cancer (CRC) research. The integration of EpCAM-directed nanotechnology in drug delivery systems has emerged as a pivotal innovation in CRC treatment. Moreover, developing chimeric antigen receptor (CAR) T-cell and CAR natural killer (NK) cell therapies opens promising therapeutic avenues for EpCAM-positive CRC patients. Although preliminary, this review sets the stage for future advances. Additionally, this study advances our understanding of the role of non-coding RNAs in CRC, which may be pivotal in gene regulation and could provide insights into the molecular underpinning. The findings suggest that lncRNA, miRNA, and circRNA could serve as novel therapeutic targets or biomarkers, further enriching the landscape of CRC diagnostics and therapeutics.
Collapse
Affiliation(s)
- Xingyu Jiang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Sumeng Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Qi Liang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Yiqian Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China.
| | - Lingxiang Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China.
| |
Collapse
|
6
|
Bhanu H, Mittal R, Senapati U. Evaluation and Clinicopathological Correlation of ALDH1 in Colorectal Adenoma with Low-/High-Grade Dysplasia and Carcinoma. South Asian J Cancer 2023. [DOI: 10.1055/s-0043-1774402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
Colorectal carcinoma (CRC) stands as one of the most prevalent malignant neoplasms, carrying significant morbidity and mortality implications. Within colorectal carcinogenesis, cancer stem cells are recognized as key contributors, infusing tumors with aggressive traits, including chemoresistance. A group of enzymes known as ALDH1 exhibits stem cell properties, potentially playing a role in colorectal neoplasms. This study aims to evaluate ALDH1 expression in colonic neoplasms and its correlation with clinicopathological parameters. The research encompasses 50 consecutive cases, involving CRC (30) and colorectal adenoma (20), gathered prospectively from September 2019 to August 2021, as well as archived cases from January 2018 to August 2019. Histological examination was conducted on CRC cases to assess tumor type, grade, lymphovascular invasion, perineural invasion, mitosis, and necrosis, while colorectal adenomas were subjected to histological grading. ALDH1 immunohistochemistry was performed on both CRC and adenoma specimens. Statistical analysis utilized SPSS 20 software, employing the chi-squared test and Fischer's exact test. A higher count of adenoma cases displayed positive staining (p = 0.0005) and greater expression (p = 0.036) in comparison to carcinoma cases. The other clinicopathological parameters didn't demonstrate notable associations. Adenomas with low-grade dysplasia exhibited a higher frequency of positive ALDH1 staining and expression than those with high-grade dysplasia. In malignant cases, a higher proportion of positive staining was observed in lower-stage disease compared to higher-stage disease. The heightened staining and expression outcomes of ALDH1 in adenomas versus carcinomas, as well as their presence in lower-stage carcinomas, suggest the potential acquisition of novel mutations and the proliferation of distinct clonal stem cell subsets during disease progression. The absence of ALDH1 in adenoma/carcinoma could indicate a poorer prognosis and an increased likelihood of disease progression to a higher stage. Comprehensive multi-institutional and validation studies are needed to enhance our understanding of ALDH1's role in colorectal oncogenesis, as well as its viability as a targeted or personalized therapy option.
Collapse
Affiliation(s)
- Himanshi Bhanu
- Department of Pathology, Kalinga Institute of Medical Sciences, Patia, Bhubaneswar, Odisha, India
| | - Ruchi Mittal
- Department of Pathology, Kalinga Institute of Medical Sciences, Patia, Bhubaneswar, Odisha, India
- Department of Histopathology, Bagchi-Sri Shankara Cancer Centre and Research Institute, Bhubaneswar, Odisha, India
| | - Urmila Senapati
- Department of Pathology, Kalinga Institute of Medical Sciences, Patia, Bhubaneswar, Odisha, India
| |
Collapse
|
7
|
Ding P, Chen P, Ouyang J, Li Q, Li S. Clinicopathological and prognostic value of epithelial cell adhesion molecule in solid tumours: a meta-analysis. Front Oncol 2023; 13:1242231. [PMID: 37664060 PMCID: PMC10468606 DOI: 10.3389/fonc.2023.1242231] [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: 06/18/2023] [Accepted: 07/27/2023] [Indexed: 09/05/2023] Open
Abstract
Background Malignant tumors, mainly solid tumors, are a significant obstacle to the improvement of life expectancy at present. Epithelial cell adhesion molecule (EpCAM), a cancer stem cell biomarker, showed widespread expression in most normal epithelial cells and most cancers. Although the clinical significance of EpCAM in various malignant solid tumors has been studied extensively, the latent relationships between EpCAM and pathological and clinical characteristics in solid tumors and differences in the roles of EpCAM among tumors have not been clearly determined. The destination point of this study was to analyze the value of EpCAM in solid tumors in clinicopathological and prognostic dimension using a meta-analysis approach. Method and materials A comprehensive and systematic search of the researches published up to March 7th, 2022, in PubMed, EMBASE, Web of Science, Cochrane library and PMC databases was performed. The relationships between EpCAM overexpression, clinicopathological characteristics, and survival outcomes were analyzed. Pooled hazard ratios (HRs) with 95% confidence intervals (CIs) and odds ratios (ORs) were estimated as indicators of the degree of correlation. This research was registered on PROSPERO (International prospective register of systematic reviews), ID: CRD42022315070. Results In total, 57 articles and 14184 cases were included in this study. High EpCAM expression had a significant coherence with a poorer overall survival (OS) (HR: 1.30, 95% CI: 1.08-1.58, P < 0.01) and a worse disease-free survival (DFS) (HR: 1.58, 95% CI: 1.28-1.95, P < 0.01), especially of gastrointestinal tumors' OS (HR: 1.50, 95% CI: 1.15-1.95, P < 0.01), and DFS (HR: 1.84, 95% CI: 1.52-2.33, P < 0.01). The DFS of head and neck tumors (HR: 2.33, 95% CI: 1.51-3.61, P < 0.01) was also associated with the overexpression of EpCAM. There were no positive relationships between the overexpression of EpCAM and sex (RR: 1.03, 95% CI: 0.99-1.07, P = 0.141), T classification (RR: 0.93, 95% CI: 0.82-1.06, P = 0.293), lymph node metastasis (RR: 0.85, 95% CI: 0.54-1.32, P = 0.461), distant metastasis (RR: 0.97, 95% CI: 0.84-1.10, P = 0.606), vascular infiltration (RR: 1.05, 95% CI: 0.85-1.29, P = 0.611), and TNM stage (RR: 0.93, 95% CI: 0.83-1.04, P = 0.187). However, the overexpression of EpCAM exhibited a significant association with the histological grades (RR: 0.88, 95% CI: 0.80-0.97, P < 0.01). Conclusion Based on pooled HRs, the positive expression of EpCAM was totally correlated to a worse OS and DFS in solid tumors. The expression of EpCAM was related to a worse OS in gastrointestinal tumors and a worse DFS in gastrointestinal tumors and head and neck tumors. Moreover, EpCAM expression was correlated with the histological grade. The results presented pointed out that EpCAM could serve as a prognostic biomarker for gastrointestinal and head and neck tumors. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier CRD42022315070.
Collapse
Affiliation(s)
- Peiwen Ding
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Panyu Chen
- Operating Room, Sichuan University West China Hospital School of Nursing, Chengdu, China
| | - Jiqi Ouyang
- Department of Gastroenterology, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Qiang Li
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shijie Li
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Demirci NS, Çavdar E, Erdem GU, Hatipoglu E, Celik E, Sezer S, Yolcu A, Dogan M, Seber ES. Is the serum level of survivin, an antiapoptotic protein, a potential predictive and prognostic biomarker in metastatic pancreatic cancer? Medicine (Baltimore) 2023; 102:e34014. [PMID: 37352081 PMCID: PMC10289789 DOI: 10.1097/md.0000000000034014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/15/2023] [Accepted: 05/25/2023] [Indexed: 06/25/2023] Open
Abstract
In the present study, we aimed to assess the association between the serum survivin level and overall survival and treatment response rates in metastatic pancreatic cancer (MPC). Serum samples were prospectively collected from 41 patients with newly diagnosed MPC patients and 41 healthy individuals (control group) to assess the survivin levels. The median survivin level was 136.2 ng/mL in patients with MPC and 52 ng/mL in healthy individuals (P = .028). Patients were divided into low- and high-survivin groups according to the baseline median survivin level. Patients with a high serum survivin level compared with a low serum survivin level had shorter median progression-free survival (2.39 vs 7.06 months; P = .008, respectively) and overall survival (3.74 vs 9.52 months; P = .026, respectively). Patients with higher serum survivin levels had significantly worse response rates (P = .007). The baseline high level of serum survivin in patients with MPC may be associated with treatment resistance and poor prognosis. A confirmation will be needed for these results in future large multicenter prospective studies.
Collapse
Affiliation(s)
- Nebi Serkan Demirci
- Department of Medical Oncology, Faculty of Medicine, Istanbul University-Cerrahpasa Cerrahpasa, Turkey
| | - Eyyüp Çavdar
- Department of Oncology, Faculty of Medicine, Tekirdag Namik Kemal University, Turkey
| | - Gokmen Umut Erdem
- Department of Medical Oncology, Başakşehir Çam and Sakura City Hospital, Turkey
| | - Engin Hatipoglu
- Department of General Surgery, Faculty of Medicine, Istanbul University-Cerrahpasa Cerrahpasa, Turkey
| | - Emir Celik
- Department of Medical Oncology, Haydarpaşa Numune Training and Research Hospital, University of Health Sciences, Turkey
| | - Sevilay Sezer
- Department of Biochemistry, Ministry of Health Ankara City Hospital, Turkey
| | - Ahmet Yolcu
- Department of Radiation Oncology, Tekirdag Namik Kemal University Faculty of Medicine, Turkey
| | - Mutlu Dogan
- Department of Medical Oncology, Ankara Oncology Training and Research Hospital, Turkey
| | - Erdogan Selcuk Seber
- Department of Oncology, Faculty of Medicine, Tekirdag Namik Kemal University, Turkey
| |
Collapse
|
9
|
Giacomelli M, Monti M, Pezzola DC, Lonardi S, Bugatti M, Missale F, Cioncada R, Melocchi L, Giustini V, Villanacci V, Baronchelli C, Manenti S, Imberti L, Giurisato E, Vermi W. Immuno-Contexture and Immune Checkpoint Molecule Expression in Mismatch Repair Proficient Colorectal Carcinoma. Cancers (Basel) 2023; 15:3097. [PMID: 37370706 DOI: 10.3390/cancers15123097] [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: 05/05/2023] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Colorectal carcinoma (CRC) represents a lethal disease with heterogeneous outcomes. Only patients with mismatch repair (MMR) deficient CRC showing microsatellite instability and hyper-mutated tumors can obtain clinical benefits from current immune checkpoint blockades; on the other hand, immune- or target-based therapeutic strategies are very limited for subjects with mismatch repair proficient CRC (CRCpMMR). Here, we report a comprehensive typing of immune infiltrating cells in CRCpMMR. We also tested the expression and interferon-γ-modulation of PD-L1/CD274. Relevant findings were subsequently validated by immunohistochemistry on fixed materials. CRCpMMR contain a significantly increased fraction of CD163+ macrophages (TAMs) expressing TREM2 and CD66+ neutrophils (TANs) together with decrease in CD4-CD8-CD3+ double negative T lymphocytes (DNTs); no differences were revealed by the analysis of conventional and plasmacytoid dendritic cell populations. A fraction of tumor-infiltrating T-cells displays an exhausted phenotype, co-expressing PD-1 and TIM-3. Remarkably, expression of PD-L1 on fresh tumor cells and TAMs was undetectable even after in vitro stimulation with interferon-γ. These findings confirm the immune suppressive microenvironment of CRCpMMR characterized by dense infiltration of TAMs, occurrence of TANs, lack of DNTs, T-cell exhaustion, and interferon-γ unresponsiveness by host and tumor cells. Appropriate bypass strategies should consider these combinations of immune escape mechanisms in CRCpMMR.
Collapse
Affiliation(s)
- Mauro Giacomelli
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Diego Cesare Pezzola
- Department of Surgery, Surgery Division II, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Silvia Lonardi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Mattia Bugatti
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Francesco Missale
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Department of Head & Neck Oncology & Surgery Otorhinolaryngology, Antoni Van Leeuwenhoek-Nederlands Kanker Instituut, 1066 CX Amsterdam, The Netherlands
| | - Rossella Cioncada
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Laura Melocchi
- Department of Pathology, Fondazione Poliambulanza, 25124 Brescia, Italy
| | - Viviana Giustini
- CREA Laboratory, AIL Center for Hemato-Oncologic Research, Diagnostic Department, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Vincenzo Villanacci
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Carla Baronchelli
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Stefania Manenti
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Luisa Imberti
- Section of Microbiology, University of Brescia, 25123 Brescia, Italy
| | - Emanuele Giurisato
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| | - William Vermi
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Department of Pathology and Immunology, School of Medicine, Washington University, Saint Louis, MO 63130, USA
| |
Collapse
|
10
|
Cloning, Expression, and Purification of the Human Synthetic Survivin Protein in Escherichia coli Using Response Surface Methodology (RSM). Mol Biotechnol 2023; 65:326-336. [PMID: 34564769 DOI: 10.1007/s12033-021-00399-4] [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: 07/06/2021] [Accepted: 09/13/2021] [Indexed: 02/07/2023]
Abstract
Survivin is one of the novel members of the apoptosis inhibitor protein family in humans. The main activity of the Survivin protein is to suppress caspases activity resulting in negative regulation of apoptosis. Survivin protein can be a potential target for the treatment of cancers between cancerous and normal cells. In the present research, the synthetic Survivin gene with PelB secretion signal peptide was cloned into a prokaryotic expression vector pET21a. The recombinant plasmid pET21a-PelB-Surv was expressed in Escherichia coli (E.coli) BL21, and the relative molecular mass of expressed protein was calculated 34,000 g/mol, approximately. The recombinant protein was purified through chromatography column and characterized by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Response surface methodology (RSM) was used to design 20 experiments for optimization of IPTG concentration, post-induction period, and cell density of induction (OD600). The optimum levels of the selected parameters were successfully determined to be 0.28 mM for IPTG concentration, 10 h for post-induction period, and 3.40768 for cell density (OD600). These findings resulted in 4.14-fold increases in the Survivin production rate of optimum expression conditions (93.6363 mg/ml).
Collapse
|
11
|
Application of plasma membrane proteomics to identify cancer biomarkers. Proteomics 2023. [DOI: 10.1016/b978-0-323-95072-5.00008-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
12
|
Hubbard JM, Tőke ER, Moretto R, Graham RP, Youssoufian H, Lőrincz O, Molnár L, Csiszovszki Z, Mitchell JL, Wessling J, Tóth J, Cremolini C. Safety and Activity of PolyPEPI1018 Combined with Maintenance Therapy in Metastatic Colorectal Cancer: an Open-Label, Multicenter, Phase Ib Study. Clin Cancer Res 2022; 28:2818-2829. [PMID: 35472243 PMCID: PMC9365360 DOI: 10.1158/1078-0432.ccr-22-0112] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/11/2022] [Accepted: 04/25/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE Although chemotherapy is standard of care for metastatic colorectal cancer (mCRC), immunotherapy has no role in microsatellite stable (MSS) mCRC, a "cold" tumor. PolyPEPI1018 is an off-the-shelf, multi-peptide vaccine derived from 7 tumor-associated antigens (TAA) frequently expressed in mCRC. This study assessed PolyPEPI1018 combined with first-line maintenance therapy in patients with MSS mCRC. PATIENTS AND METHODS Eleven patients with MSS mCRC received PolyPEPI1018 and Montanide ISA51VG adjuvant subcutaneously, combined with fluoropyrimidine/biologic following first-line induction with chemotherapy and a biologic (NCT03391232). In Part A of the study, 5 patients received a single dose; in Part B, 6 patients received up to three doses of PolyPEPI1018 every 12 weeks. The primary objective was safety; secondary objectives were preliminary efficacy, immunogenicity at peripheral and tumor level, and immune correlates. RESULTS PolyPEPI1018 vaccination was safe and well tolerated. No vaccine-related serious adverse event occurred. Eighty percent of patients had CD8+ T-cell responses against ≥3 TAAs. Increased density of tumor-infiltrating lymphocytes were detected post-treatment for 3 of 4 patients' liver biopsies, combined with increased expression of immune-related gene signatures. Three patients had objective response according to RECISTv1.1, and 2 patients qualified for curative surgery. Longer median progression-free survival for patients receiving multiple doses compared with a single dose (12.5 vs. 4.6 months; P = 0.017) suggested a dose-efficacy correlation. The host HLA genotype predicted multi-antigen-specific T-cell responses (P = 0.01) indicative of clinical outcome. CONCLUSIONS PolyPEPI1018 added to maintenance chemotherapy for patients with unresectable, MSS mCRC was safe and associated with specific immune responses and antitumor activity warranting further confirmation in a randomized, controlled setting.
Collapse
Affiliation(s)
| | - Enikő R Tőke
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - Roberto Moretto
- Department of Translational Research and New Technologies in Medicine and Surgery, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | | | | | - Orsolya Lőrincz
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - Levente Molnár
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - Zsolt Csiszovszki
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | | | | | - József Tóth
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| |
Collapse
|
13
|
Al‐Qahtani SM, Gadalla SE, Guo M, Ericsson C, Hägerstrand D, Nistér M. The association between Annexin A2 and epithelial cell adhesion molecule in breast cancer cells. Cancer Rep (Hoboken) 2022; 5:e1498. [PMID: 34240826 PMCID: PMC9124509 DOI: 10.1002/cnr2.1498] [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: 03/05/2021] [Revised: 06/05/2021] [Accepted: 06/14/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND The epithelial cell adhesion molecule (EpCAM) is a type I transmembrane and glycosylated protein, which is overexpressed in many neoplasms. However, EpCAM has no known ligand partners and the mechanisms by which it functions are not fully understood. AIM This study was performed to discover novel partners of EpCAM, which may provide a better understanding of its functions. METHODS The membrane fraction of the ERα+ noninvasive breast cancer cell line ZR-75-1 and MCF-7 was extracted and followed by co-immunoprecipitation of EpCAM using C-10, a mouse monoclonal antibody raised against amino acids 24-93 of the EpCAM molecule. As a negative control, MDA-MB-231 and Hs578T were used since they express a negligible amount of EpCAM and are known as EpCAM-/low ERα-/low invasive and tumorigenic breast cancer cell lines. RESULTS Annexin A2 (ANXA2) was found to be selectively and differentially co-immunoprecipitated with EpCAM in the ERα+ breast cancer cells MCF-7 and ZR-75-1. ANXA2 is a multifunctional protein and known to act as a co-receptor for tissue plasminogen activator (tPA) on the surface of endothelial and cancer cells, thereby affecting fibrinolytic activity and neoangiogenesis as well as invasive and metastatic properties. In this study, the association between EpCAM and ANXA2 was found to affect the activity of tPA. CONCLUSION This study concludes that ANXA2 co-localizes with EpCAM at the plasma membrane, and the co-localization may have functional implications. Data suggest that EpCAM supports ANXA2 to function as a co-receptor for the tPA, and that EpCAM has a regulatory function on the expression and subcellular localization of ANXA2.
Collapse
Affiliation(s)
- Saad Misfer Al‐Qahtani
- Department of Oncology‐PathologyKarolinska InstitutetStockholmSweden
- Department of Pathology, College of Medicine and Najran University HospitalNajran UniversityNajranSaudi Arabia
| | | | - Min Guo
- Department of Oncology‐PathologyKarolinska InstitutetStockholmSweden
| | | | | | - Monica Nistér
- Department of Oncology‐PathologyKarolinska InstitutetStockholmSweden
| |
Collapse
|
14
|
Tumor-suppressive role of Smad ubiquitination regulatory factor 2 in patients with colorectal cancer. Sci Rep 2022; 12:5495. [PMID: 35361871 PMCID: PMC8971512 DOI: 10.1038/s41598-022-09390-8] [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: 08/12/2021] [Accepted: 03/23/2022] [Indexed: 12/24/2022] Open
Abstract
Smad ubiquitination regulatory factor 2 (Smurf2) plays various roles in cancer progression. However, the correlation between Smurf2 and clinical outcomes has not been determined in patients diagnosed with colorectal cancer and colorectal liver metastases. We analyzed 66 patients with colorectal cancer who developed liver metastases. Smurf2 expression was assessed using immunohistochemical analysis of primary and metastatic liver tumors. High Smurf2 expression in both primary and metastatic tumors was significantly associated with longer overall survival time and time to surgical failure. Multivariate analyses revealed that low Smurf2 expression in primary tumors was an independent predictor of poor prognosis. In vitro experiments using colon cancer cell lines demonstrated that short interfering RNA knockdown of Smurf2 increased cell migration and tumor sphere formation. Western blot analyses revealed that Smurf2 knockdown increased the protein expression of epithelial cell adhesion molecule (EpCAM). Thus, in summary, high Smurf2 expression in cancer cells was found to be an independent predictor of better prognosis in patients with primary colorectal cancer and consequent liver metastases. The tumor-suppressive role of Smurf2 was found to be associated with cell migration and EpCAM expression; hence, Smurf2 can be considered a positive biomarker of cancer stem cell-like properties.
Collapse
|
15
|
The Expression and Prognostic Value of Cancer Stem Cell Markers, NRF2, and Its Target Genes in TAE/TACE-Treated Hepatocellular Carcinoma. Medicina (B Aires) 2022; 58:medicina58020212. [PMID: 35208536 PMCID: PMC8879934 DOI: 10.3390/medicina58020212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/26/2022] [Accepted: 01/26/2022] [Indexed: 11/25/2022] Open
Abstract
Background and Objectives: Activation of NRF2, a key transcription factor of cytoprotectant against oxidative stress, and its target genes are associated with aggressive tumor progression, metastasis and poor survival. In addition, NRF2 signaling mediates cancer stem cell (CSC)-like properties in hepatocellular carcinoma (HCC) cells. Moreover, CSCs have been associated with HCC onset and unfavorable prognosis. Transcatheter arterial embolization (TAE) and/or transcatheter arterial chemoembolization (TACE), which attempt to restrict blood supply to diminish tumor growth, can create a hypoxic environment. However, its effect on NRF2 signaling and CSC marker CD133 in the context of prognosis of HCCs have not been investigated. Therefore, we studied the possible role of the expressions of NRF2, its target genes and CSC markers CD133 and EpCAM on the survival of HCC patients after TAE/TACE. Materials and Methods: RT-qPCR was performed with 120 tumor (T) and adjacent tumor (N) tissue pairs. Expression of a single marker or combination was assessed for associations with survival of HCC patients after TAE/TACE. Results: The result of multivariate Cox regression showed that vascular invasion (HR, 1.821; p = 0.015), metastasis (HR, 2.033; p = 0.049) and CD133 overexpression (HR, 2.013; p = 0.006) were associated with poor survival. In a Kaplan–Meier survival analysis, patients with high expression of CD133 had shorter overall survival (OS) than those with low expression of CD133 in post-TAE/TACE HCC (p < 0.001). In contrast, neither NRF2 nor components of its signaling pathway correlated with survival. Combination marker analysis showed that co-expression of NQO1 and CD133 was associated with poor outcome. Conclusions: This study suggests that analyzing the expression status of CD133 alone and co-expression of NQO1 and CD133 may have additional value in predicting the outcome of TAE/TACE-treated HCC patients.
Collapse
|
16
|
Kalantari E, Taheri T, Fata S, Abolhasani M, Mehrazma M, Madjd Z, Asgari M. Significant co-expression of putative cancer stem cell markers, EpCAM and CD166, correlates with tumor stage and invasive behavior in colorectal cancer. World J Surg Oncol 2022; 20:15. [PMID: 35016698 PMCID: PMC8751119 DOI: 10.1186/s12957-021-02469-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/02/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The crucial oncogenic role of cancer stem cells (CSCs) in tumor maintenance, progression, drug resistance, and relapse has been clarified in different cancers, particularly in colorectal cancer (CRC). The current study was conducted to evaluate the co-expression pattern and clinical significance of epithelial cell adhesion molecules (EpCAM) and activated leukocyte cell adhesion (CD166 or ALCAM) in CRC patients. METHODS This study was carried out on 458 paraffin-embedded CRC specimens by immunohistochemistry on tissue microarray (TMA) slides. RESULTS Elevated expression of EpCAM and CD166 was observed in 61.5% (246/427) and 40.5% (164/405) of CRC cases. Our analysis showed a significant positive association of EpCAM expression with tumor size (P = 0.02), tumor stage (P = 0.007), tumor differentiate (P = 0.005), vascular (P = 0.01), neural (P = 0.01), and lymph node (P = 0.001) invasion. There were no significant differences between CD166 expression and clinicopathological parameters. Moreover, the combined analysis demonstrated a reciprocal significant correlation between EpCAM and CD166 expression (P = 0.02). Interestingly, there was a significant positive correlation between EpCAM/CD166 phenotypes expression and tumor stage (P = 0.03), tumor differentiation (P = 0.05), neural, and lymph node invasion (P =0.01). CONCLUSIONS The significant correlation of EpCAM and CD166 expression and their association with tumor progression and aggressive behavior is the reason for the suggestion of these two CSC markers as promising targets to promote novel effective targeted-therapy strategies for cancer treatment in the present study.
Collapse
Affiliation(s)
- Elham Kalantari
- Oncopathology Research Center, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
| | - Tahereh Taheri
- Department of Pathology, Iran University of Medical Sciences, Tehran, Iran
| | - Saba Fata
- Department of Pathology, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Abolhasani
- Oncopathology Research Center, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
- Department of Pathology, Hasheminejad kidney Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mitra Mehrazma
- Oncopathology Research Center, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran.
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mojgan Asgari
- Oncopathology Research Center, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran.
- Department of Pathology, Hasheminejad kidney Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Afshari AR, Mollazadeh H, Henney NC, Jamialahmad T, Sahebkar A. Effects of statins on brain tumors: a review. Semin Cancer Biol 2021; 73:116-133. [DOI: 10.1016/j.semcancer.2020.08.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/04/2020] [Accepted: 08/09/2020] [Indexed: 02/06/2023]
|
18
|
Li Y, Lu W, Yang J, Edwards M, Jiang S. Survivin as a biological biomarker for diagnosis and therapy. Expert Opin Biol Ther 2021; 21:1429-1441. [PMID: 33877952 DOI: 10.1080/14712598.2021.1918672] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Survivin (SVN) is a member of the inhibitor of apoptosis (IAP) protein family that promotes cellular proliferation and inhibits apoptosis. Overexpression of SVN is associated with autoimmune disease, hyperplasia, and tumors and can be used as a biomarker in these diseases. SVN is widely recognized as a tumor-associated antigen (TAA) and has become an important target for cancer diagnosis and treatment.Areas covered: We reviewed SVN research progress from the PubMed and clinical trials focused on SVN from https://clinicaltrials.gov since 2000 and anticipate future developments in the field. The trials reviewed cover various modalities including diagnostics for early detection and disease progression, small molecule inhibitors of the SVN pathway and immunotherapy targeting SVN epitopes.Expert opinion: The most promising developments involve anti-SVN immunotherapy, with several therapeutic SVN vaccines under evaluation in phase I/II trials. SVN is an important new immune-oncology target that expands the repertoire of individualized combination treatments for cancer.
Collapse
Affiliation(s)
- Yuming Li
- Department of Oncology, University of Oxford, Oxford, UK.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Wenshu Lu
- Department of Oncology, University of Oxford, Oxford, UK
| | - Jiarun Yang
- Department of Oncology, University of Oxford, Oxford, UK
| | - Mark Edwards
- Department of Research and Development, Oxford Vacmedix UK Ltd, Oxford, UK
| | - Shisong Jiang
- Department of Oncology, University of Oxford, Oxford, UK.,Department of Research and Development, Oxford Vacmedix UK Ltd, Oxford, UK
| |
Collapse
|
19
|
Hollandsworth HM, Turner MA, Hoffman RM, Bouvet M. A review of tumor-specific fluorescence-guided surgery for colorectal cancer. Surg Oncol 2021; 36:84-90. [PMID: 33316684 PMCID: PMC7855598 DOI: 10.1016/j.suronc.2020.11.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 11/18/2020] [Accepted: 11/27/2020] [Indexed: 01/08/2023]
Abstract
The present study reviews the use of tumor-specific antibodies conjugated to fluorescent dyes in preclinical and clinical studies to enhance visualization of primary tumors and metastases for fluorescence-guided surgery (FGS) in colorectal cancer (CRC). A search strategy was developed using the peer-reviewed National Center for Biotechnology Information (NCBI) database on PubMed. Studies using tumor-specific fluorescence imaging and FGS techniques on murine models of colorectal cell lines or patient-derived orthotopic xenograft (PDOX) colorectal cancer are reviewed. A total of 24 articles were identified that met the inclusion criteria, 21 preclinical and 3 clinical trials. The most widely used target antigen in preclinical and clinical trials was carcinoembryonic antigen (CEA). Mouse studies and clinical studies have demonstrated that the use of FGS in CRC can aid in decreased residual tumor and decreased rates of recurrence. As the mainstay of colorectal cancer treatment is surgery, the addition of intraoperative fluorescence imaging can help locate tumor margins, visualize occult micro-metastases, drive surgical decision making and improve patient outcomes.
Collapse
Affiliation(s)
- Hannah M Hollandsworth
- Department of Surgery, University of California San Diego, San Diego, CA, USA; Moores Cancer Center, University of California San Diego, San Diego, CA, USA
| | - Michael A Turner
- Department of Surgery, University of California San Diego, San Diego, CA, USA; Moores Cancer Center, University of California San Diego, San Diego, CA, USA
| | - Robert M Hoffman
- Department of Surgery, University of California San Diego, San Diego, CA, USA; Moores Cancer Center, University of California San Diego, San Diego, CA, USA; AntiCancer Inc., San Diego, CA, USA; VA San Diego Healthcare System, San Diego, CA, USA
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, CA, USA; Moores Cancer Center, University of California San Diego, San Diego, CA, USA; VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
20
|
cIAP2 via NF-κB signalling affects cell proliferation and invasion in hepatocellular carcinoma. Life Sci 2020; 266:118867. [PMID: 33310033 DOI: 10.1016/j.lfs.2020.118867] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/24/2022]
Abstract
AIMS To investigate the role of cIAP2 in the malignant biological behaviours of hepatocellular carcinoma (HCC) cells and determine its mechanism of action. MAIN METHODS cIAP2 protein expression was detected via immunohistochemistry (IHC) in 102 HCC specimens and 43 paracancerous liver tissues, and its relationship with clinicopathological features and patient prognosis was analysed. Then, short interfering RNA (siRNA) technology was used to knock down cIAP2 expression in BEL7402 and HepG2 cells. Cell Counting Kit-8 (CCK8) and Transwell assays were used to determine cell proliferation and invasion after knockdown of cIAP2 expression. The relationship between cIAP2 and the NF-κB pathway was explored via western blotting (WB) and a dual luciferase reporter system. Finally, nude mouse models of liver cancer were established to detect the effect of cIAP2 on tumourigenicity and the proliferation activity of orthotopic HCC cells. KEY FINDINGS cIAP2 expression was significantly increased in HCC tissues and was correlated with intravascular thrombosis in HCC. High cIAP2 expression was correlated with poor patient prognosis. cIAP2 knockdown significantly reduced the proliferation and invasion of BEL7402 and HepG2 cells and the activity of the NF-κB pathway. Animal experiments showed that cIAP2 knockdown reduced the tumourigenicity of HepG2 cells in the liver of nude mice and the proliferation activity of the orthotopic HCC cells. SIGNIFICANCE cIAP2 plays an important role in HCC proliferation and invasion and may exert its effects via the NF-κB signalling pathway.
Collapse
|
21
|
Rezaee M, Gheytanchi E, Madjd Z, Mehrazma M. Clinicopathological Significance of Tumor Stem Cell Markers ALDH1 and CD133 in Colorectal Carcinoma. IRANIAN JOURNAL OF PATHOLOGY 2020; 16:40-50. [PMID: 33391379 PMCID: PMC7691712 DOI: 10.30699/ijp.2020.127441.2389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/23/2020] [Indexed: 12/15/2022]
Abstract
Background & Objective: Colorectal cancer (CRC) is the third most common cancer worldwide with a high mortality rate. The main causes of death in patients are recurrence and metastasis which are mainly attributed to the small subpopulation of cells within tumors called cancer stem cells (CSCs). This study aimed to evaluate the correlation between the expression of ALDH1 and CD133 as CSC associated markers and clinicopathological characteristics in CRC. Methods: In this cross-sectional study, a total of 483 CRC tumor samples were immunohistochemically stained for detection of CD133 and ALDH1 markers. Correlations of marker expression with clinicopathological factors were also evaluated. Results: There was a significant correlation between the luminal intensity of CD133 and neural invasion (P=0.05) and between the cytoplasmic intensity of CD133 and metastasis (P=0.05). In terms of H-score, a positive significant relation was observed between cytoplasmic expression of CD133 and lymph node (P=0.02), neural (P=0.04) and vascular invasion (P=0.02). The ALDH1 cytoplasmic expression showed a significant correlation with tumor size (P=0.001). Conclusion: Our findings showed that increased expression of CD133 and ALDH1 is associated with tumor progression and worse outcomes in CRC patients. These markers can be good candidates for localized targeting of CSCs using antibodies. Future researches need to be improved approaches for early detection of CRC, and treatment monitoring for CRC and other cancers.
Collapse
Affiliation(s)
- Maryam Rezaee
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Elmira Gheytanchi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mitra Mehrazma
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Pathology, Hasheminejad Kidney Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Vermani L, Kumar R, Kannan RR, Deka MK, Talukdar A, Kumar NS. Expression pattern of ALDH1, E-cadherin, Vimentin and Twist in early and late onset sporadic colorectal cancer. Biomark Med 2020; 14:1371-1382. [PMID: 33064013 DOI: 10.2217/bmm-2020-0206] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/30/2020] [Indexed: 12/21/2022] Open
Abstract
Aim: To evaluate the expression pattern of ALDH1 (aldehyde dehydrogenase 1), E-cadherin, Vimentin and Twist in early and late onset sporadic colorectal cancer (CRC) and to study association of their expression with the occurrence of CRC at a young age. Materials & methods: Immunohistochemistry of ALDH1, E-cadherin, Vimentin and Twist was performed on 103 pretreated CRC biopsy samples. Results: ALDH1 expression was found to have strong correlation with early onset CRC (p < 0.0001). Conclusion: High ALDH1 expression correlates with the early onset of CRC. ALDH1 over-expression correlates with poor overall survival in colon cancer.
Collapse
Affiliation(s)
- Litika Vermani
- Mizoram University, Biotechnology, Aizawl, Mizoram 796004, India
| | - Rajeev Kumar
- Cachar Cancer Hospital & Research Centre, Research Silchar, 788015, India
| | | | - Monoj K Deka
- Silchar Medical College & Hospital, 788015, India
| | - Anuradha Talukdar
- Cachar Cancer Hospital & Research Centre, Research Silchar, 788015, India
| | | |
Collapse
|
23
|
Eslami-S Z, Cortés-Hernández LE, Alix-Panabières C. Epithelial Cell Adhesion Molecule: An Anchor to Isolate Clinically Relevant Circulating Tumor Cells. Cells 2020; 9:cells9081836. [PMID: 32764280 PMCID: PMC7464831 DOI: 10.3390/cells9081836] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
In the last few decades, the epithelial cell adhesion molecule (EpCAM) has received increased attention as the main membrane marker used in many enrichment technologies to isolate circulating tumor cells (CTCs). Although there has been a great deal of progress in the implementation of EpCAM-based CTC detection technologies in medical settings, several issues continue to limit their clinical utility. The biology of EpCAM and its role are not completely understood but evidence suggests that the expression of this epithelial cell-surface protein is crucial for metastasis-competent CTCs and may not be lost completely during the epithelial-to-mesenchymal transition. In this review, we summarize the most significant advantages and disadvantages of using EpCAM as a marker for CTC enrichment and its potential biological role in the metastatic cascade.
Collapse
|
24
|
Pádua D, Figueira P, Ribeiro I, Almeida R, Mesquita P. The Relevance of Transcription Factors in Gastric and Colorectal Cancer Stem Cells Identification and Eradication. Front Cell Dev Biol 2020; 8:442. [PMID: 32626705 PMCID: PMC7314965 DOI: 10.3389/fcell.2020.00442] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
Gastric and colorectal cancers have a high incidence and mortality worldwide. The presence of cancer stem cells (CSCs) within the tumor mass has been indicated as the main reason for tumor relapse, metastasis and therapy resistance, leading to poor overall survival. Thus, the elimination of CSCs became a crucial goal for cancer treatment. The identification of these cells has been performed by using cell-surface markers, a reliable approach, however it lacks specificity and usually differs among tumor type and in some cases even within the same type. In theory, the ideal CSC markers are those that are required to maintain their stemness features. The knowledge that CSCs exhibit characteristics comparable to normal stem cells that could be associated with the expression of similar transcription factors (TFs) including SOX2, OCT4, NANOG, KLF4 and c-Myc, and signaling pathways such as the Wnt/β-catenin, Hedgehog (Hh), Notch and PI3K/AKT/mTOR directed the attention to the use of these similarities to identify and target CSCs in different tumor types. Several studies have demonstrated that the abnormal expression of some TFs and the dysregulation of signaling pathways are associated with tumorigenesis and CSC phenotype. The disclosure of common and appropriate biomarkers for CSCs will provide an incredible tool for cancer prognosis and treatment. Therefore, this review aims to gather the new insights in gastric and colorectal CSC identification specially by using TFs as biomarkers and divulge promising drugs that have been found and tested for targeting these cells.
Collapse
Affiliation(s)
- Diana Pádua
- i3S – Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Paula Figueira
- i3S – Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Inês Ribeiro
- i3S – Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Raquel Almeida
- i3S – Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Biology, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Patrícia Mesquita
- i3S – Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| |
Collapse
|
25
|
Tsunedomi R, Yoshimura K, Suzuki N, Hazama S, Nagano H. Clinical implications of cancer stem cells in digestive cancers: acquisition of stemness and prognostic impact. Surg Today 2020; 50:1560-1577. [PMID: 32025858 DOI: 10.1007/s00595-020-01968-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/14/2020] [Indexed: 02/06/2023]
Abstract
Digestive system cancers are the most frequent cancers worldwide and often associated with poor prognosis because of their invasive and metastatic characteristics. Recent studies have found that the plasticity of cancer cells can impart cancer stem-like properties via the epithelial-mesenchymal transition (EMT). Cancer stem-like properties such as tumor initiation are integral to the formation of metastasis, which is the main cause of poor prognosis. Numerous markers of cancer stem cells (CSCs) have been identified in many types of cancer. Therefore, CSCs, via their stem cell-like functions, may play an important role in prognosis after surgery. While several reports have described prognostic analysis using CSC markers, few reviews have summarized CSCs and their association with prognosis. Herein, we review the prognostic potential of eight CSC markers, CD133, CD44, CD90, ALDH1A1, EPCAM, SOX2, SOX9, and LGR5, in digestive cancers including those of the pancreas, colon, liver, gastric, and esophagus.
Collapse
Affiliation(s)
- Ryouichi Tsunedomi
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| | - Kiyoshi Yoshimura
- Showa University Clinical Research Institute for Clinical Pharmacology and Therapeutics, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Nobuaki Suzuki
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shoichi Hazama
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.,Faculty of Medicine, Department of Translational Research and Developmental Therapeutics against Cancer, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| |
Collapse
|
26
|
Cha BS, Park KS, Park JS. Signature mRNA markers in extracellular vesicles for the accurate diagnosis of colorectal cancer. J Biol Eng 2020; 14:4. [PMID: 32042310 PMCID: PMC7001337 DOI: 10.1186/s13036-020-0225-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/21/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND With the increasing incidence of colorectal cancer (CRC), its accurate diagnosis is critical and in high demand. However, conventional methods are not ideal due to invasiveness and low accuracy. Herein, we aimed to identify efficient CRC mRNA markers in a non-invasive manner using CRC-derived extracellular vesicles (EVs). The expression levels of EV mRNAs from cancer cell lines were compared with those of a normal cell line using quantitative polymerase chain reaction. Eight markers were evaluated in plasma EVs from CRC patients and healthy controls. The diagnostic value of each marker, individually or in combination, was then determined using recessive operating characteristics analyses and the Mann-Whitney U test. RESULTS Eight mRNA markers (MYC, VEGF, CDX2, CD133, CEA, CK19, EpCAM, and CD24) were found to be more abundant in EVs derived from cancer cell lines compared to control cell lines. A combination of VEGF and CD133 showed the highest sensitivity (100%), specificity (80%), and accuracy (93%) and an area under the curve of 0.96; hence, these markers were deemed to be the CRC signature. Moreover, this signature was found to be highly expressed in CRC-derived EVs compared to healthy controls. CONCLUSIONS VEGF and CD133 mRNAs comprise a unique CRC signature in EVs that has the potential to act as a novel, non-invasive, and accurate biomarker that would improve the current diagnostic platform for CRC, while also serving to strengthen the value of EV mRNA as diagnostic markers for myriad of diseases.
Collapse
Affiliation(s)
- Byung Seok Cha
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, Republic of Korea
| | - Ki Soo Park
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, Republic of Korea
| | - Jun Seok Park
- School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Colorectal Cancer Center, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| |
Collapse
|
27
|
Yang J, Isaji T, Zhang G, Qi F, Duan C, Fukuda T, Gu J. EpCAM associates with integrin and regulates cell adhesion in cancer cells. Biochem Biophys Res Commun 2020; 522:903-909. [DOI: 10.1016/j.bbrc.2019.11.152] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022]
|
28
|
Mohamed SY, Kaf RM, Ahmed MM, Elwan A, Ashour HR, Ibrahim A. The Prognostic Value of Cancer Stem Cell Markers (Notch1, ALDH1, and CD44) in Primary Colorectal Carcinoma. J Gastrointest Cancer 2019; 50:824-837. [PMID: 30136202 DOI: 10.1007/s12029-018-0156-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Cancer stem cells proved to have a vital role in cell migration, invasion, metastasis, and treatment resistance of colorectal cancer (CRC) that subsequently lead to poor clinical outcomes. These stem cells may be a novel therapeutic target for the management of CRC progression. Signals of the Notch-1 pathway are responsible for acquisition of stem cell characters. ALDH1 and CD44 are usually detected in stem cells in colorectal cancer. AIM The aims of this work are to evaluate the immunohistochemical expression of cancer stem cell markers ALDH1, Notch1, and CD44 in colorectal cancer and investigate their correlation with clinicopathological characters and patient survival. METHODS Paraffin-embedded specimens of 70 patients with primary colorectal carcinoma were analyzed for Notch 1, ALDH1, and CD44 expressions by immunohistochemistry. RESULTS Notch1 was mainly located in the cytoplasm of CRC tissues, rarely expressed in adjacent normal tissues. A highly statistically significant relationship was found between grading, lymphovascular invasion, the degree of lymphocytic infiltration, peritumoral budding, lymph node ratio, lymph node metastasis, and Notch1 expression (p < 0.001). There was a highly statistically significant relationship found between AJCC stage and Notch1 expression (p < 0.001). CD44 was mainly located in the cell membrane of CRC tissues. A highly statistically significant relationship was found between grading (p = 0.006), lymphovascular invasion, the degree of lymphocytic infiltration, peritumoral budding, lymph node metastasis, lymph node ratio, and CD44 expression (p < 0.001). There was a highly statistically significant relationship found between AJCC stage and CD44 expression (p < 0.001). ALDH1 was detected in the cytoplasm of the CRC tissue. A highly statistically significant relationship was found between grading, lymphovascular invasion, the degree of lymphocytic infiltration, peritumoral budding, lymph node metastasis, lymph node ratio, and ALDH1 expression (p < 0.001). There was a highly statistically significant relationship found between AJCC stage and ALDH1 expression (p < 0.001). There is a highly statistically significant direct correlation between Notch1, CD44 expression, and ALDH1 expression (p < 0.001). CONCLUSIONS There is a substantial correlation between Notch 1, ALDH1, and CD44 as cancer stem cell markers and lymph node metastasis, advanced stage and tumor recurrence in colorectal carcinoma. CONCLUSION Expression of stem cell markers ALDH1, Notch1, and CD44 correlates with poor prognosis in a CRC and represents an independent prognostic factor. They are associated with a feature of epithelial-mesenchymal transition evidenced by their association with high tumor burden.
Collapse
Affiliation(s)
- Salem Y Mohamed
- Gastroenterology and Hepatology Unit, Internal Medicine Department, Faculty of Medicine, Zagazig University, Faqous city, Markaz El-Zakazik, Ash Sharqia Governorate, 44519, Egypt.
| | - Randa Mohamed Kaf
- Department of Pathology, Faculty of Medicine, Zagazig University, Markaz El-Zakazik, Ash Sharqia Governorate, 44519, Egypt
| | - Mona Mostafa Ahmed
- Department of Pathology, Faculty of Medicine, Zagazig University, Markaz El-Zakazik, Ash Sharqia Governorate, 44519, Egypt
| | - Amira Elwan
- Department of Clinical Oncology, Faculty of Medicine, Zagazig University, Markaz El-Zakazik, Ash Sharqia Governorate, 44519, Egypt
| | - Hassan R Ashour
- Department of General Surgery, Faculty of Medicine, Zagazig University, Markaz El-Zakazik, Ash Sharqia Governorate, 44519, Egypt
| | - Amr Ibrahim
- Department of General Surgery, Faculty of Medicine, Zagazig University, Markaz El-Zakazik, Ash Sharqia Governorate, 44519, Egypt
| |
Collapse
|
29
|
Xu S, Song J, Zhang ZH, Fu L, Gao L, Xie DD, Yu DX, Xu DX, Sun GP. The Vitamin D status is associated with serum C-reactive protein and adhesion molecules in patients with renal cell carcinoma. Sci Rep 2019; 9:16719. [PMID: 31723229 PMCID: PMC6853912 DOI: 10.1038/s41598-019-53395-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022] Open
Abstract
Low vitamin D status is associated with an increased risk of renal cell carcinoma (RCC). This study investigated the association of vitamin D status with serum C-reactive protein (CRP) and adhesion molecules among RCC patients. Fifty newly diagnosed RCC patients and 100 age- and sex-matched controls were recruited. As expected, serum 25(OH)D level was lower in RCC patients than in controls. By contrast, serum levels of CRP, an inflammatory molecule, and ICAM, LAMA4 and EpCAM, three adhesion molecules, were higher in RCC patients than in controls. All RCC patients were divided into two groups: H-VitD (>20 ng/ml) or L-VitD (<20 ng/ml). Interestingly, the levels of serum CRP and all adhesion molecules were higher in RCC patients with L-VitD than those with H-VitD. Nuclear vitamin D receptor (VDR) was downregulated and nuclear factor kappa B (NF-κB) was activated in cancerous tissues. The in vitro experiments found that VitD3 suppressed NF-κB activation and adhesion molecules in RCC cells. Moreover, VitD3 suppressed NF-κB through reinforcing physical interaction between VDR and NF-κB p65 subunit in RCC cells. These results provide a mechanistic explanation for the association among low vitamin D status, local inflammation and increased expression of adhesion molecules among RCC patients.
Collapse
Affiliation(s)
- Shen Xu
- Department of Oncology, First Affiliated Hospital, Anhui Medical University, Hefei, 230022, China
| | - Jin Song
- Department of Urology, Second Affiliated Hospital, Anhui Medical University, Hefei, 230601, China
| | - Zhi-Hui Zhang
- Department of Urology, Second Affiliated Hospital, Anhui Medical University, Hefei, 230601, China
| | - Lin Fu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China.,Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - Lan Gao
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China.,Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - Dong-Dong Xie
- Department of Urology, Second Affiliated Hospital, Anhui Medical University, Hefei, 230601, China
| | - De-Xin Yu
- Department of Urology, Second Affiliated Hospital, Anhui Medical University, Hefei, 230601, China
| | - De-Xiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China. .,Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China.
| | - Guo-Ping Sun
- Department of Oncology, First Affiliated Hospital, Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
30
|
Tsai SL, Chang YC, Sarvagalla S, Wang S, Coumar MS, Cheung CHA. Cloning, expression, and purification of the recombinant pro-apoptotic dominant-negative survivin T34A-C84A protein in Escherichia coli. Protein Expr Purif 2019; 160:73-83. [DOI: 10.1016/j.pep.2019.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 02/14/2019] [Accepted: 04/12/2019] [Indexed: 01/07/2023]
|
31
|
Mahmood NA, Abdulghany ZS, Al-Sudani IM. Expression of Aldehyde Dehydrogenase (ALDH1) and ATP Binding Cassette Transporter G2 (ABCG2) in Iraqi Patients with Colon Cancer and the Relation with Clinicopathological Features. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2019; 7:234-240. [PMID: 31516883 PMCID: PMC6709934 DOI: 10.22088/ijmcm.bums.7.4.234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/25/2019] [Indexed: 01/16/2023]
Abstract
Tumor initiation cells or cancer stem cell markers ABCG2 and ALDH1 play pivotal roles in invasion, metastasis and resistance to cytotoxic agents. In this study, we evaluated the expression levels of ALDH1 and ABCG2 in Iraqi patients with colon cancer and/or benign colon tumors. We also investigated the association between the expression levels of these markers and patient clincopathological features. The expression levels of ALDH1 and ABCG2 in cancer tissues as well as in benign tumor samples were determined by immunohistochemistry using tumor tissues microarray of TNM (Tumor, Node, Metastasis) in 42 patients with colon cancer samples as well as in 18 corresponding benign tumors. Immunohistochemistry showed that ALDH1 and ABCG2 expression levels increased to 80% and 76%, respectively in colon cancer tissues as compared to 33% and 28% in benign tumor tissues. The expression levels of ALDH1 and ABCG2 were associated with tumor stages. No significant association was found between the expressions levels of these markers and tumor size, gender, patients' age, and lymph node involvement. These results indicate that the expression levels of ALDH1 and ABCG2 increased in colon cancer tissues compared to benign tumor tissues in Iraqi patients.
Collapse
Affiliation(s)
- Noah Abdulqader Mahmood
- Molecular Biology Department, Iraqi Center for Cancer and Medical Genetics Research, Mustansiriyah University, Baghdad, Iraq.
| | - Zaynab Saad Abdulghany
- Molecular Biology Department, Iraqi Center for Cancer and Medical Genetics Research, Mustansiriyah University, Baghdad, Iraq.
| | | |
Collapse
|
32
|
Sandberg TP, Sweere I, van Pelt GW, Putter H, Vermeulen L, Kuppen PJ, Tollenaar RAEM, Mesker WE. Prognostic value of low CDX2 expression in colorectal cancers with a high stromal content - a short report. Cell Oncol (Dordr) 2019; 42:397-403. [PMID: 30847807 DOI: 10.1007/s13402-019-00436-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2019] [Indexed: 01/07/2023] Open
Abstract
PURPOSE Lack of expression of the intestinal transcription factor CDX2 in colorectal cancer (CRC) identifies patients with a poor prognosis. This biomarker has previously been suggested to be prognostic in CRCs with a high stromal content based on mRNA expression data. We investigated the prognostic value of CDX2 expression in microsatellite stable CRC stratified by stromal content using microscopy-based techniques. METHODS AND RESULTS The study included a cohort of 236 patients with stage I-IV CRC. We assessed by microscopy the tumour-stroma ratio (TSR) and the immunohistochemical CDX2 intensity. We found that patients of the stroma-high group had a worse prognosis compared to those of the stroma-low group [disease-free survival in a multivariate analysis (DFSmultivariate) HR 1.52 (95% CI 1.05-2.21)]. In our cohort, low CDX2 expression (14.6%) showed prognostic value for DFSmultivariate [HR 1.93 (95% CI 1.16-3.23)]. Interestingly, when stratifying the cohort by TSR, no prognostic difference was observed related to CDX2 expression in stroma-low tumours. However, CDX2 expression was found to be prognostic within the stroma-high group [DFSmultivariate HR 3.02 (95% CI 1.49-6.13)]. The p value for interaction between TSR and CDX2 status was borderline significant in DFS (p = 0.071). CONCLUSIONS The present study confirms a poor outcome of patients with stroma-high tumours. Low CDX2 expression in tumours with a high stromal content identified patients with a particularly poor prognosis. The present study did not reveal a clear difference in TSR associated with CDX2 status and survival. This method, solely based on microscopy, identifies patients who have a high risk of relapse and a poor outcome, and who may benefit from targeted therapy.
Collapse
Affiliation(s)
- Tessa P Sandberg
- Department of Surgery, Leiden University Medical Centrum, Albinusdreef 2, 2333ZA, Leiden, The Netherlands. .,Department of Pathology, Leiden University Medical Centrum, Leiden, The Netherlands.
| | - Iris Sweere
- Department of Pathology, Leiden University Medical Centrum, Leiden, The Netherlands
| | - Gabi W van Pelt
- Department of Surgery, Leiden University Medical Centrum, Albinusdreef 2, 2333ZA, Leiden, The Netherlands
| | - Hein Putter
- Department of Medical Statistics, Leiden University Medical Centrum, Leiden, The Netherlands
| | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center & Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Peter J Kuppen
- Department of Surgery, Leiden University Medical Centrum, Albinusdreef 2, 2333ZA, Leiden, The Netherlands
| | - Rob A E M Tollenaar
- Department of Surgery, Leiden University Medical Centrum, Albinusdreef 2, 2333ZA, Leiden, The Netherlands
| | - Wilma E Mesker
- Department of Surgery, Leiden University Medical Centrum, Albinusdreef 2, 2333ZA, Leiden, The Netherlands
| |
Collapse
|
33
|
Yahyazadeh Mashhadi SM, Kazemimanesh M, Arashkia A, Azadmanesh K, Meshkat Z, Golichenari B, Sahebkar A. Shedding light on the EpCAM: An overview. J Cell Physiol 2019; 234:12569-12580. [DOI: 10.1002/jcp.28132] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/30/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Seyed Muhammad Yahyazadeh Mashhadi
- Department of Virology Pasteur Institute of Iran Tehran Iran
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Production Expert at Samandaroo 8 (Biotech Pharmaceutical) Co. Mashhad Iran
| | | | - Arash Arashkia
- Department of Virology Pasteur Institute of Iran Tehran Iran
| | | | - Zahra Meshkat
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences Mashhad Iran
| | - Behrouz Golichenari
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
| | - Amirhosein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
- Neurogenic inflammation Research Center, Mashhad University of Medical Sciences Mashhad Iran
- School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
34
|
Trinh A, Lädrach C, Dawson HE, Ten Hoorn S, Kuppen PJK, Reimers MS, Koopman M, Punt CJA, Lugli A, Vermeulen L, Zlobec I. Tumour budding is associated with the mesenchymal colon cancer subtype and RAS/RAF mutations: a study of 1320 colorectal cancers with Consensus Molecular Subgroup (CMS) data. Br J Cancer 2018; 119:1244-1251. [PMID: 30385823 PMCID: PMC6251036 DOI: 10.1038/s41416-018-0230-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 07/16/2018] [Accepted: 07/20/2018] [Indexed: 01/03/2023] Open
Abstract
Background Tumour budding is an important prognostic factor in colorectal cancer (CRC). Molecular profiling of tumour buds suggests (partial) epithelial–mesenchymal transition and cancer stem-cell phenotype, similarly described in the “mesenchymal” Consensus Molecular Subtype 4 (CMS4), which identifies a particularly poor prognostic subgroup. Here, we determine the association of tumour budding with CMS classification, prognosis, and response to therapy. Methods AMC-AJCCII-90 cohort (n = 76, stage II) was evaluated for peritumoural budding on H&E slides. LUMC (n = 270, stage I–IV), CAIRO (n = 504, metastatic CRC) and CAIRO2 (n = 472, metastatic CRC) cohorts were investigated for intratumoural budding using pan-cytokeratin-stained tissue microarrays. Budding was scored as count/area, then classified as <5 or ≥5 buds. For all cohorts, CMS classifications were available (gene-expression/immunohistochemistry-based classifiers). Results High (≥5) budding predicted a worse outcome in multivariate analysis in AMC-AJCCII-90 (p = 0.018), LUMC (p < 0.0001), and CAIRO (p = 0.03), and in CAIRO2 (continuous variable, p = 0.02). Tumour budding counts were higher in CMS4 compared to epithelial CMS2/3 cancers (p < 0.01, all), and associated with KRAS/BRAF mutations (p < 0.01, AMC-AJCCII-90, CAIRO, CAIRO2). Conclusion Tumour budding is an adverse prognostic factor across all CRC stages and is associated with the mesenchymal CMS4 phenotype. KRAS/BRAF mutations are strongly correlated with tumour budding suggesting their involvement in the regulation of this process.
Collapse
Affiliation(s)
- Anne Trinh
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Claudia Lädrach
- Institute of Pathology, University of Bern, Bern, Switzerland
| | | | - Sanne Ten Hoorn
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Marlies S Reimers
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Miriam Koopman
- Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cornelis J A Punt
- Medical Oncology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Inti Zlobec
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
35
|
Lee SJ, Chung KY, Kwon JE, Yoon SO, Kim SK. Expression of EpCAM in adenoid cystic carcinoma. Pathology 2018; 50:737-741. [PMID: 30389218 DOI: 10.1016/j.pathol.2018.08.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/20/2018] [Accepted: 08/23/2018] [Indexed: 01/14/2023]
Abstract
The mutational landscape of adenoid cystic carcinoma (ACC) is currently being revealed, but further studies are needed to identify biomarkers as therapeutic targets or prognostic factors of ACC. In this study, we investigated the expression of epithelial cell adhesion molecule (EpCAM) in ACCs. We retrospectively collected 83 cases of surgically resected ACCs. Using tissue microarray, we conducted immunohistochemical staining using the anti-EpCAM antibody. EpCAM expression was analysed by intensity score and the total immunostaining score. The positivity was 97.6% (81/83 cases), regardless of the intensity score. A higher histological grade (p = 0.006) and specific tumour location (non-salivary gland origin, p = 0.02) showed a correlation with higher EpCAM intensity. Higher EpCAM expression by total immunostaining score was associated with histological grade (p = 0.004), distant metastasis (p = 0.004) and poorer prognosis (overall survival p = 0.015 and progression-free survival p = 0.033). We suggest EpCAM as a candidate prognostic marker and a putative therapeutic target in ACC. Also, ACCs arising from salivary gland and non-salivary gland sites, respectively, might display different pathophysiologies in which EpCAM could play a role.
Collapse
Affiliation(s)
- Seok Joo Lee
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Kee Yang Chung
- Department of Dermatology, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji Eun Kwon
- Department of Pathology, Ajou University School of Medicine, Suwon, South Korea
| | - Sun Och Yoon
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Kyum Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
36
|
Aberrantly Expressed Genes and miRNAs in Slow Transit Constipation Based on RNA-Seq Analysis. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2617432. [PMID: 30186855 PMCID: PMC6112260 DOI: 10.1155/2018/2617432] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/26/2018] [Accepted: 07/18/2018] [Indexed: 02/06/2023]
Abstract
Background This study aims to identify the key genes and miRNAs in slow transit constipation (STC). Methods MRNA and miRNA expression profiling were obtained. Differentially expressed genes (DEGs) and miRNAs were identified followed by the regulatory network construction. Functional annotation analysis and protein-protein interaction (PPI) network were conducted. The electronic validation was performed. Results Hsa-miR-2116-3p, hsa-miR-3622a-5p, hsa-miR-424-5p, and hsa-miR-1273-3p covered most DEGs. HLA-DRB1, HLA-DRB5, C3, and ICAM were significantly involved in staphylococcus aureus infection. The PPI network generated several hub proteins including ZBTB16, FBN1, CCNF, and CDK1. Electronic validation of HLA-DRB1, PTGDR, MKI67, BIRC5, CCNF, and CDK1 was consistent with the RNA-sequencing analysis. Conclusion Our study might be helpful in understanding the pathology of STC at the molecular level.
Collapse
|
37
|
Jiménez G, Hackenberg M, Catalina P, Boulaiz H, Griñán-Lisón C, García MÁ, Perán M, López-Ruiz E, Ramírez A, Morata-Tarifa C, Carrasco E, Aguilera M, Marchal JA. Mesenchymal stem cell's secretome promotes selective enrichment of cancer stem-like cells with specific cytogenetic profile. Cancer Lett 2018; 429:78-88. [PMID: 29733965 DOI: 10.1016/j.canlet.2018.04.042] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 04/27/2018] [Accepted: 04/27/2018] [Indexed: 12/15/2022]
|
38
|
Herreros-Pomares A, Aguilar-Gallardo C, Calabuig-Fariñas S, Sirera R, Jantus-Lewintre E, Camps C. EpCAM duality becomes this molecule in a new Dr. Jekyll and Mr. Hyde tale. Crit Rev Oncol Hematol 2018; 126:52-63. [DOI: 10.1016/j.critrevonc.2018.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 02/08/2023] Open
|
39
|
Tamatani T, Takamaru N, Ohe G, Akita K, Nakagawa T, Miyamoto Y. Expression of CD44, CD44v9, ABCG2, CD24, Bmi-1 and ALDH1 in stage I and II oral squamous cell carcinoma and their association with clinicopathological factors. Oncol Lett 2018; 16:1133-1140. [PMID: 29963189 DOI: 10.3892/ol.2018.8703] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 04/11/2018] [Indexed: 12/28/2022] Open
Abstract
Cancer stem cells (CSCs) exhibit self-replication, self-differentiation, drug resistance and immune evasion activities. In recent years CSCs have become increasingly important for the treatment of malignant tumors. CSCs express specific markers, including cluster of differentiation (CD)44, CD44 variant 9 (CD44v9), ATP-binding cassette sub-family G member 2 (ABCG2), CD24, B lymphoma Mo-MLV insertion region 1 homolog (BMI-1) and aldehyde dehydrogenase 1 (ALDH1). However, the prognostic value of their expression in patients with oral squamous cell carcinoma (OSCC) are not well known. The present study evaluated these markers in stage I and II patients with OSCC and examined the association between T classification, histological differentiation, classification of invasion mode, lymph node metastasis and disease-free survival rate. Tissue specimens were obtained from 70 patients with stage I or II OSCC following either surgery or biopsy. Immunohistochemistry was performed and positive staining was defiend as 10% positive cells. CD44 and CD44v9 expressions were strongly detected in all OSCC tissues compared with normal epithelial cells. A total of 22 (31.4%) cases expressed ABCG2 and there was a significant association between ABCG2 expression and invasion. A total of 41 cases (59.0%) expressed CD24 and there was a significant association between CD24 expression and invasion. A total of 33 cases (47.1%) expressed BMI-1 and there was a significant association between BMI-1 expression and the disease-free survival rate. A total of 18 cases (25.7%) expressed ALDH1. Although there was no association between ALDH1 expression and T classification, there were significant associations between ALDH1 expression and histological differentiation, invasion mode, metastasis and the disease-free survival rate. Multivariate analysis revealed that ALDH1 expression was the only prognostic factor for disease-free survival rate. The results of the present study suggest that the positivity of ALDH1 detected in patients with OSCC correlates with the number of cells undergoing epithelial mesenchymal transition and metastasis. These findings indicated that the expression of ALDH1 may be an effective prognostic marker indicating the survival of patients with stage I and II OSCC.
Collapse
Affiliation(s)
- Tetsuya Tamatani
- Department of Oral Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan
| | - Natsumi Takamaru
- Department of Oral Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan
| | - Go Ohe
- Department of Oral Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan
| | - Kazuya Akita
- Department of Oral Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan
| | - Takayuki Nakagawa
- Department of Oral Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan
| | - Youji Miyamoto
- Department of Oral Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan
| |
Collapse
|
40
|
Kim JH, Bae JM, Song YS, Cho NY, Lee HS, Kang GH. Clinicopathologic, molecular, and prognostic implications of the loss of EPCAM expression in colorectal carcinoma. Oncotarget 2017; 7:13372-87. [PMID: 26528695 PMCID: PMC4924648 DOI: 10.18632/oncotarget.5618] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 10/19/2015] [Indexed: 01/26/2023] Open
Abstract
We aimed to comprehensively investigate the clinicopathologic and molecular implications of altered epithelial cell adhesion molecule (EPCAM) expression in colorectal carcinoma (CRC). EPCAM immunohistochemical expression, EPCAM 3' end deletion, EPCAM promoter methylation, microsatellite instability (MSI), and the CpG island methylator phenotype (CIMP) were analyzed in large cohorts of human CRCs. Among 218 MSI-high CRCs, complete loss (CL) of EPCAM expression was observed in two cases, both of which displayed MSH2 deficiency and EPCAM 3' deletion. Thirty-one of the 218 MSI-high CRCs demonstrated the partial loss (PL) of EPCAM expression without EPCAM deletion or methylation and were correlated with CIMP-high and poor disease-free survival. Histologically, foci exhibiting EPCAM loss in EPCAM-PL tumors were dominantly distributed in poorly differentiated tumor components and/or in the invasive tumor front. The implications of EPCAM-PL were further validated in a consecutive series of 726 CRCs. EPCAM-PL (n = 50; 6.9%) was also associated with CIMP-high and adverse pathologic factors and was confirmed to be an independent poor prognostic factor in CRC (HR, 1.57; 95% CI, 1.04 to 2.39). EPCAM-CL can be used to screen for EPCAM deletion-induced Lynch syndrome-associated CRC, whereas EPCAM-PL can be used as an indicator of tumor aggressiveness and poor prognosis in CRC.
Collapse
Affiliation(s)
- Jung Ho Kim
- Department of Pathology, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Jeong Mo Bae
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Young Seok Song
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Nam-Yun Cho
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Gyeong Hoon Kang
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea.,Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
41
|
Miller T, McCoy M, Hemmings C, Bulsara M, Iacopetta B, Platell C. The prognostic value of cancer stem-like cell markers SOX2 and CD133 in stage III colon cancer is modified by expression of the immune-related markers FoxP3, PD-L1 and CD3. Pathology 2017; 49:721-730. [DOI: 10.1016/j.pathol.2017.08.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 08/07/2017] [Accepted: 08/14/2017] [Indexed: 01/19/2023]
|
42
|
TAB3 upregulates Survivin expression to promote colorectal cancer invasion and metastasis by binding to the TAK1-TRAF6 complex. Oncotarget 2017; 8:106565-106576. [PMID: 29290971 PMCID: PMC5739756 DOI: 10.18632/oncotarget.22497] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/28/2017] [Indexed: 12/23/2022] Open
Abstract
Transforming growth factor-β-activated kinase 1 (TAK1)-binding protein 3 (TAB3) is involved in cancer proliferation and metastasis, but its role in colorectal cancer remains unclear. In this study, we demonstrated that TAB3 is upregulated in colorectal cancer tissues and that high TAB3 levels correlated with tumor metastasis and a poor prognosis in colorectal cancer. In addition, TAB3 knockdown decreased Survivin expression and suppressed colorectal cancer cell migration and invasion in vitro, and reduced liver metastasis in vivo. Importantly, we found that TAB3 regulated Survivin expression by activating the NF-κB pathway through the formation of the TAK1-TAB3-TRAF6 complex. These findings suggest TAB3 may be a useful prognostic biomarker in colorectal cancer and a target for treatment of metastatic colorectal cancer.
Collapse
|
43
|
Histone Chaperone ASF1A Predicts Poor Outcomes for Patients With Gastrointestinal Cancer and Drives Cancer Progression by Stimulating Transcription of β-Catenin Target Genes. EBioMedicine 2017. [PMID: 28625518 PMCID: PMC5514402 DOI: 10.1016/j.ebiom.2017.06.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Epigenetic mechanisms play a key role in gastrointestinal cancer (GIC) development and progression, and most studies have been focused on aberrant DNA methylation and histone modifying enzymes. However, the histone H3–H4 chaperone ASF1A is an important factor regulating chromatin assembling and gene transcription, while it is currently unclear whether ASF1A is involved in cancer pathogenesis. The present study is thus designed to address this issue. Here we showed that ASF1A expression was widespread in GIC-derived cell lines and up-regulated in primary GIC. Higher levels of ASF1A expression predicted significantly shorter patient overall survival in colorectal cancer (P = 0.0012). The further analyses of the GEO dataset validate higher ASF1A expression as a prognostic factor for CRC patients. Mechanistically, ASF1A interacted with β-catenin and promoted the transcription of β-catenin target genes including c-MYC, cyclin D1, ZEB1 and LGR5, thereby stimulating proliferation, stemness and migration/invasion of GIC cells. β-Catenin inhibition abolished these effects of ASF1A. Moreover, the ASF1A-β-catenin-ZEB1 axis down-regulated E-Cadherin expression, thereby contributing to enhanced migration/invasion of GIC cells. ASF1A over-expression and depletion facilitated and inhibited in vivo tumor growth and/or metastasis in mouse xenograft models, respectively. Taken together, ASF1A is aberrantly over-expressed in GIC tumors and plays key roles in GIC development and progression by stimulating the transcription of β-catenin target genes. ASF1A may thus be a novel target for GIC therapy and a potential prognostic marker. ASF1A is over-expressed in GIC and predicts poor patient outcomes. ASF1A interacts with β-catenin, facilitates the expression of its targets and promotes stemness and invasion of GIC cells. Targeting ASF1A could be a novel epigenetic strategy against GIC.
Gastrointestinal cancer (GIC) is the leading cause of cancer-related death worldwide and invasion or distant metastasis account for the majority of mortalities due to the limited treatment choices. We determined the effect of ASF1A, the histone H3–H4 chaperone, on GIC pathogenesis. We found that ASF1A expression was up-regulated in primary GIC and higher ASF1A levels predicted significantly shorter patient overall survival in colorectal cancer. ASF1A interacted with β-catenin and promoted the transcription of β-catenin target genes, thereby stimulating proliferation, stemness and migration/invasion of GIC cells. ASF1A may thus be a novel target for GIC therapy and a useful prognostic marker.
Collapse
|
44
|
Wahab SR, Islam F, Gopalan V, Lam AKY. The Identifications and Clinical Implications of Cancer Stem Cells in Colorectal Cancer. Clin Colorectal Cancer 2017; 16:93-102. [DOI: 10.1016/j.clcc.2017.01.011] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 11/16/2016] [Accepted: 01/13/2017] [Indexed: 12/18/2022]
|
45
|
Han S, Zong S, Shi Q, Li H, Liu S, Yang W, Li W, Hou F. Is Ep-CAM Expression a Diagnostic and Prognostic Biomarker for Colorectal Cancer? A Systematic Meta-Analysis. EBioMedicine 2017; 20:61-69. [PMID: 28558958 PMCID: PMC5478257 DOI: 10.1016/j.ebiom.2017.05.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/11/2017] [Accepted: 05/22/2017] [Indexed: 01/15/2023] Open
Abstract
Background Cancer stem cell (CSC) epithelial cell adhesion molecule (Ep-CAM) is frequently expressed in colorectal cancer (CRC). However, the clinical significance of Ep-CAM expression in CRC is not clear. This study evaluated whether Ep-CAM provided valuable insight as a molecular biomarker for CRC diagnosis and prognosis and the potential of Ep-CAM as a novel therapeutic target in CRC. Methods Publications were selected online using electronic databases. The pooled odds ratios (ORs) or hazard ratios (HRs) with their 95% confidence intervals (95% CIs), and the combined sensitivity, specificity, and area under the curve (AUC) were calculated and summarized. Results Eleven eligible articles published in English involving 4561 cases were analyzed in this study. Ep-CAM expression was significantly higher in CRC compared with normal controls, and its overexpression was negatively linked to tumor differentiation, tumor stage, vascular invasion, depth of tumor invasion, lymph node metastasis, distant metastasis, and tumor budding in CRC patients. The loss of Ep-CAM expression positively correlated with these characteristics. Multivariate analysis of loss of Ep-CAM expression correlated with a poor prognosis in disease-free survival (DFS), disease-specific survival (DSS), and overall survival (OS). The pooled sensitivity, specificity and AUC values of Ep-CAM expression in patients with CRC vs. normal controls were 0.93, 0.90, and 0.94, respectively. Conclusions The present findings suggest that Ep-CAM expression may be associated with CRC carcinogenesis, while the loss of Ep-CAM expression is correlated with the progression, metastasis, and poor prognosis of CRC. Ep-CAM expression may be a useful biomarker for the clinical diagnosis of CRC. Cancer stem cell (CSC) epithelial cell adhesion molecule (Ep-CAM) expression may correlate with CRC tumorigenesis. Frequent overexpression of Ep-CAM was a favorable factor for CRC progression and metastasis.
Loss of Ep-CAM expression correlated with the progression, metastasis, and poor prognosis of patients with CRC. Ep-CAM expression may be a potential marker for the detection of CRC.
Ep-CAM expression was reported in CRC, but no clear direction for the diagnostic and prognostic effects of Ep-CAM expression was documented in patients with CRC. We performed a systematic meta-analysis of the existing evidence to determine the clinical significance of Ep-CAM expression in CRC. The findings indicated that Ep-CAM expression was associated with CRC risk. Frequent overexpression of Ep-CAM correlated with a decreased risk of CRC progression and metastasis, and loss of Ep-CAM expression played an important role in CRC progression, metastasis and prognosis. The detection of Ep-CAM expression may be a promising biomarker in diagnosing CRC.
Collapse
Affiliation(s)
- Susu Han
- Oncology Department of Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai TCM University, 274 Zhijiang Road, Shanghai 200071, People's Republic of China
| | - Shaoqi Zong
- Oncology Department of Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai TCM University, 274 Zhijiang Road, Shanghai 200071, People's Republic of China
| | - Qi Shi
- Oncology Department of Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai TCM University, 274 Zhijiang Road, Shanghai 200071, People's Republic of China
| | - Hongjia Li
- Oncology Department of Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai TCM University, 274 Zhijiang Road, Shanghai 200071, People's Republic of China
| | - Shanshan Liu
- Oncology Department of Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai TCM University, 274 Zhijiang Road, Shanghai 200071, People's Republic of China
| | - Wei Yang
- Oncology Department of Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai TCM University, 274 Zhijiang Road, Shanghai 200071, People's Republic of China
| | - Wen Li
- Oncology Department of Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai TCM University, 274 Zhijiang Road, Shanghai 200071, People's Republic of China.
| | - Fenggang Hou
- Oncology Department of Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai TCM University, 274 Zhijiang Road, Shanghai 200071, People's Republic of China.
| |
Collapse
|
46
|
Gao S, Sun Y, Liu X, Zhang D, Yang X. EpCAM and COX-2 expression are positively correlated in human breast cancer. Mol Med Rep 2017; 15:3755-3760. [DOI: 10.3892/mmr.2017.6447] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 02/09/2017] [Indexed: 11/06/2022] Open
|
47
|
Trinh A, Trumpi K, De Sousa E Melo F, Wang X, de Jong JH, Fessler E, Kuppen PJK, Reimers MS, Swets M, Koopman M, Nagtegaal ID, Jansen M, Hooijer GKJ, Offerhaus GJA, Kranenburg O, Punt CJ, Medema JP, Markowetz F, Vermeulen L. Practical and Robust Identification of Molecular Subtypes in Colorectal Cancer by Immunohistochemistry. Clin Cancer Res 2017; 23:387-398. [PMID: 27459899 DOI: 10.1158/1078-0432.ccr-16-0680] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 06/23/2016] [Accepted: 07/15/2016] [Indexed: 12/22/2022]
Abstract
PURPOSE Recent transcriptomic analyses have identified four distinct molecular subtypes of colorectal cancer with evident clinical relevance. However, the requirement for sufficient quantities of bulk tumor and difficulties in obtaining high-quality genome-wide transcriptome data from formalin-fixed paraffin-embedded tissue are obstacles toward widespread adoption of this taxonomy. Here, we develop an immunohistochemistry-based classifier to validate the prognostic and predictive value of molecular colorectal cancer subtyping in a multicenter study. EXPERIMENTAL DESIGN Tissue microarrays from 1,076 patients with colorectal cancer from four different cohorts were stained for five markers (CDX2, FRMD6, HTR2B, ZEB1, and KER) by immunohistochemistry and assessed for microsatellite instability. An automated classification system was trained on one cohort using quantitative image analysis or semiquantitative pathologist scoring of the cores as input and applied to three independent clinical cohorts. RESULTS This classifier demonstrated 87% concordance with the gold-standard transcriptome-based classification. Application to three validation datasets confirmed the poor prognosis of the mesenchymal-like molecular colorectal cancer subtype. In addition, retrospective analysis demonstrated the benefit of adding cetuximab to bevacizumab and chemotherapy in patients with RAS wild-type metastatic cancers of the canonical epithelial-like subtypes. CONCLUSIONS This study shows that a practical and robust immunohistochemical assay can be employed to identify molecular colorectal cancer subtypes and uncover subtype-specific therapeutic benefit. Finally, the described tool is available online for rapid classification of colorectal cancer samples, both in the format of an automated image analysis pipeline to score tumor core staining, and as a classifier based on semiquantitative pathology scoring. Clin Cancer Res; 23(2); 387-98. ©2016 AACR.
Collapse
Affiliation(s)
- Anne Trinh
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kari Trumpi
- Cancer Center UMC Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Felipe De Sousa E Melo
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Molecular Oncology, Genentech Inc., South San Francisco, California
| | - Xin Wang
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Joan H de Jong
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Evelyn Fessler
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Marlies S Reimers
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Marloes Swets
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Miriam Koopman
- Cancer Center UMC Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Iris D Nagtegaal
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marnix Jansen
- Department of Pathology, Lymphoma and Myeloma Center Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Centre for Tumour Biology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gerrit K J Hooijer
- Department of Pathology, Lymphoma and Myeloma Center Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - George J A Offerhaus
- Cancer Center UMC Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Onno Kranenburg
- Cancer Center UMC Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cornelis J Punt
- Department of Medical Oncology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Florian Markowetz
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
48
|
Holah NS, Aiad HAES, Asaad NY, Elkhouly EA, Lasheen AG. Evaluation of the Role of ALDH1 as Cancer Stem Cell Marker in Colorectal Carcinoma: An Immunohistochemical Study. J Clin Diagn Res 2017; 11:EC17-EC23. [PMID: 28273973 PMCID: PMC5324418 DOI: 10.7860/jcdr/2017/22671.9291] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/14/2016] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Colorectal Carcinoma (CRC) is the third most commonly diagnosed cancer in males. Stem Cells (SC) may be involved in tumour growth, including colon cancer. Aldehyde Dehydrogenase 1 (ALDH1) is a detoxifying enzyme that might modulate SC proliferation. AIMS To evaluate the immunohistochemical expression of ALDH1 as stem cell marker in the pathogenesis of colorectal carcinoma. MATERIALS AND METHODS This retrospective study included 71 colorectal specimens (49 colorectal carcinoma, 13 adenoma and 9 normal cases) that were collected from Pathology Department, Faculty of Medicine, Menoufia University during the period from 2011 to 2015. All cases were stained by ALDH 1 antibody. Survival data were available for 31cases. RESULTS There was a statistical significant association between epithelial positivity of ALDH1 and younger age (p=0.003), right sided tumour (p=0.038), presence of lymph node invasion (p= 0.04), ulcerating gross picture (p=0.01) and presence of vascular invasion (p=0.05). Moreover, there was statistical significant association between stromal positivity of ALDH1 and smaller tumour size (p=0.03) and inverse association between stromal expression of ALDH1 and grade of tumour (p=0.000) and perineural invasion (p= 0.05). Furthermore, there was an inverse significant relation between CD44 and ALDH1 expression (p=0.001). Univariate recurrence free survival analysis revealed the bad prognostic impact of high grade (p=0.03) and female sex (p=0.02) on patient outcome. CONCLUSION Epithelial expression of ALDH1 might be associated with poor prognosis while its stromal expression might be associated with good prognosis.
Collapse
Affiliation(s)
- Nanis Shawky Holah
- Department of Pathology, Faculty of Medicine, Menoufia University, Shebien ElKom, Menoufia, Egypt
| | - Hayam Abd-El-Samie Aiad
- Professor, Department of Pathology, Faculty of Medicine, Menoufia University, Shebien ElKom, Menoufia, Egypt
| | - Nancy Yousif Asaad
- Professor, Department of Pathology, Faculty of Medicine, Menoufia University, Shebien ElKom, Menoufia, Egypt
| | - Enas Abobakr Elkhouly
- Department of Oncology, Faculty of Medicine, Menoufia University, Shebien ElKom, Menoufia, Egypt
| | - Ayat Gamal Lasheen
- Department of Pathology, Faculty of Medicine, Menoufia University, Shebien ElKom, Menoufia, Egypt
| |
Collapse
|
49
|
van Driel PBAA, Boonstra MC, Prevoo HAJM, van de Giessen M, Snoeks TJA, Tummers QRJG, Keereweer S, Cordfunke RA, Fish A, van Eendenburg JDH, Lelieveldt BPF, Dijkstra J, van de Velde CJH, Kuppen PJK, Vahrmeijer AL, Löwik CWGM, Sier CFM. EpCAM as multi-tumour target for near-infrared fluorescence guided surgery. BMC Cancer 2016; 16:884. [PMID: 27842504 PMCID: PMC5109830 DOI: 10.1186/s12885-016-2932-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 10/30/2016] [Indexed: 01/08/2023] Open
Abstract
Background Evaluation of resection margins during cancer surgery can be challenging, often resulting in incomplete tumour removal. Fluorescence-guided surgery (FGS) aims to aid the surgeon to visualize tumours and resection margins during surgery. FGS relies on a clinically applicable imaging system in combination with a specific tumour-targeting contrast agent. In this study EpCAM (epithelial cell adhesion molecule) is evaluated as target for FGS in combination with the novel Artemis imaging system. Methods The NIR fluorophore IRDye800CW was conjugated to the well-established EpCAM specific monoclonal antibody 323/A3 and an isotype IgG1 as control. The anti-EpCAM/800CW conjugate was stable in serum and showed preserved binding capacity as evaluated on EpCAM positive and negative cell lines, using flow cytometry and cell-based plate assays. Four clinically relevant orthotopic tumour models, i.e. colorectal cancer, breast cancer, head and neck cancer, and peritonitis carcinomatosa, were used to evaluate the performance of the anti-EpCAM agent with the clinically validated Artemis imaging system. The Pearl Impulse small animal imaging system was used as reference. The specificity of the NIRF signal was confirmed using bioluminescence imaging and green-fluorescent protein. Results All tumour types could clearly be delineated and resected 72 h after injection of the imaging agent. Using NIRF imaging millimetre sized tumour nodules were detected that were invisible for the naked eye. Fluorescence microscopy demonstrated the distribution and tumour specificity of the anti-EpCAM agent. Conclusions This study shows the potential of an EpCAM specific NIR-fluorescent agent in combination with a clinically validated intraoperative imaging system to visualize various tumours during surgery.
Collapse
Affiliation(s)
- P B A A van Driel
- Department of Radiology, Division of Molecular Imaging, Leiden University Medical Centre, Leiden, Netherlands.,Percuros BV, Enschede, The Netherlands
| | - M C Boonstra
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - H A J M Prevoo
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - M van de Giessen
- Department of Radiology and Division of Image Processing, Leiden University Medical Centre, Leiden, Netherlands
| | - T J A Snoeks
- Department of Radiology, Division of Molecular Imaging, Leiden University Medical Centre, Leiden, Netherlands
| | - Q R J G Tummers
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - S Keereweer
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus Medical Centre, Rotterdam, Netherlands
| | - R A Cordfunke
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, Netherlands
| | - A Fish
- Division of Biochemistry, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - B P F Lelieveldt
- Department of Radiology and Division of Image Processing, Leiden University Medical Centre, Leiden, Netherlands
| | - J Dijkstra
- Department of Radiology and Division of Image Processing, Leiden University Medical Centre, Leiden, Netherlands
| | - C J H van de Velde
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - P J K Kuppen
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands.,Antibodies for Research Applications BV, Gouda, The Netherlands
| | - A L Vahrmeijer
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - C W G M Löwik
- Department of Radiology, Division of Molecular Imaging, Leiden University Medical Centre, Leiden, Netherlands
| | - C F M Sier
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands. .,Antibodies for Research Applications BV, Gouda, The Netherlands.
| |
Collapse
|
50
|
De Rosa M, Rega D, Costabile V, Duraturo F, Niglio A, Izzo P, Pace U, Delrio P. The biological complexity of colorectal cancer: insights into biomarkers for early detection and personalized care. Therap Adv Gastroenterol 2016; 9:861-886. [PMID: 27803741 PMCID: PMC5076770 DOI: 10.1177/1756283x16659790] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancer has been ranked the third and second most prevalent of all cancers in men and women, respectively, and it represents the fourth most common cause of cancer deaths. In 2012, there were 1.4 million estimated cases of colorectal cancer worldwide, and 700,000 estimated deaths, which implies significant impact on public health, especially in economically-developed countries. In recent years, there has been an increase in the number of tumors, although this has been accompanied by decreased mortality, due to more appropriate and available information, earlier diagnosis, and improvements in treatment. Colorectal cancers are characterized by great genotypic and phenotypic heterogeneity, including tumor microenvironment and interactions between healthy and cancer cells. All of these traits confer a unique peculiarity to each tumor, which can thus be considered as an individual disease. Well conducted molecular and clinical characterization of each colorectal cancer is essential with a view to the implementation of precision oncology, and thus personalized care. This last aims at standardization of therapeutic plans chosen according to the genetic background of each specific neoplasm, to increase overall survival and reduce treatment side effects. Thus, prognostic and predictive molecular biomarkers assume a critical role in the characterization of colorectal cancer and in the determination of the most appropriate therapy.
Collapse
Affiliation(s)
- Marina De Rosa
- Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II ’, I-80131 Naples, Italy
| | - Daniela Rega
- Colorectal Surgical Oncology-Abdominal Oncology Department, Istituto Nazionale per lo Studio e la Cura dei Tumori, ‘Fondazione Giovanni Pascale’ IRCCS, I-80131 Naples, Italy
| | - Valeria Costabile
- Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II ’, I-80131 Naples, Italy
| | - Francesca Duraturo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II ’, I-80131 Naples, Italy
| | - Antonello Niglio
- Colorectal Surgical Oncology-Abdominal Oncology Department, Istituto Nazionale per lo Studio e la Cura dei Tumori, ‘Fondazione Giovanni Pascale’ IRCCS, I-80131 Naples, Italy
| | - Paola Izzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II ’, I-80131 Naples, Italy
| | - Ugo Pace
- Colorectal Surgical Oncology-Abdominal Oncology Department, Istituto Nazionale per lo Studio e la Cura dei Tumori, ‘Fondazione Giovanni Pascale’ IRCCS, I-80131 Naples, Italy
| | - Paolo Delrio
- Colorectal Surgical Oncology-Abdominal Oncology Department, Istituto Nazionale per lo Studio e la Cura dei Tumori, ‘Fondazione Giovanni Pascale’ IRCCS, I-80131 Naples, Italy
| |
Collapse
|