1
|
Anderson TS, McCormick AL, Daugherity EA, Oladejo M, Okpalanwaka IF, Smith SL, Appiah D, Wood LM, Lowe DB. Listeria-based vaccination against the pericyte antigen RGS5 elicits anti-vascular effects and colon cancer protection. Oncoimmunology 2023; 12:2260620. [PMID: 37781234 PMCID: PMC10540654 DOI: 10.1080/2162402x.2023.2260620] [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: 06/28/2023] [Accepted: 09/14/2023] [Indexed: 10/03/2023] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related mortality despite efforts to improve standard interventions. As CRC patients can benefit from immunotherapeutic strategies that incite effector T cell action, cancer vaccines represent a safe and promising therapeutic approach to elicit protective and durable immune responses against components of the tumor microenvironment (TME). In this study, we investigate the pre-clinical potential of a Listeria monocytogenes (Lm)-based vaccine targeting the CRC-associated vasculature. CRC survival and progression are reliant on functioning blood vessels to effectively mediate various metabolic processes and oxygenate underlying tissues. We, therefore, advance the strategy of initiating immunity in syngeneic mouse models against the endogenous pericyte antigen RGS5, which is a critical mediator of pathological vascularization. Overall, Lm-based vaccination safely induced potent anti-tumor effects that consisted of recruiting functional Type-1-associated T cells into the TME and reducing tumor blood vessel content. This study underscores the promising clinical potential of targeting RGS5 against vascularized tumors like CRC.
Collapse
Affiliation(s)
- Trevor S. Anderson
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Amanda L. McCormick
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Elizabeth A. Daugherity
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Izuchukwu F. Okpalanwaka
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Savanna L. Smith
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Duke Appiah
- Department of Public Health, School of Population and Public Health, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Laurence M. Wood
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Devin B. Lowe
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| |
Collapse
|
2
|
Turano M, Vicidomini R, Cammarota F, D'Agostino V, Duraturo F, Izzo P, Rosa MD. The Epithelial to Mesenchymal Transition in Colorectal Cancer Progression: The Emerging Role of Succinate Dehydrogenase Alterations and Succinate Accumulation. Biomedicines 2023; 11:biomedicines11051428. [PMID: 37239099 DOI: 10.3390/biomedicines11051428] [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: 04/01/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Colorectal cancer (CRC) stands as the third most significant contributor to cancer-related mortality worldwide. A major underlying reason is that the detection of CRC usually occurs at an advanced metastatic stage, rendering therapies ineffective. In the progression from the in situ neoplasia stage to the advanced metastatic stage, a critical molecular mechanism involved is the epithelial-to-mesenchymal transition (EMT). This intricate transformation consists of a series of molecular changes, ultimately leading the epithelial cell to relinquish its features and acquire mesenchymal and stem-like cell characteristics. The EMT regulation involves several factors, such as transcription factors, cytokines, micro RNAs and long noncoding RNAs. Nevertheless, recent studies have illuminated an emerging link between metabolic alterations and EMT in various types of cancers, including colorectal cancers. In this review, we delved into the pivotal role played by EMT during CRC progression, with a focus on highlighting the relationship between the alterations of the tricarboxylic acid cycle, specifically those involving the succinate dehydrogenase enzyme, and the activation of the EMT program. In fact, emerging evidence supports the idea that elucidating the metabolic modifications that can either induce or inhibit tumor progression could be of immense significance for shaping new therapeutic approaches and preventative measures. We conclude that an extensive effort must be directed towards research for the standardization of drugs that specifically target proteins such as SDH and SUCNR1, but also TRAP1, PDH, ERK1/2, STAT3 and the HIF1-α catabolism.
Collapse
Affiliation(s)
- Mimmo Turano
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Rosario Vicidomini
- Section on Cellular Communication, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Francesca Cammarota
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
| | - Valeria D'Agostino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Francesca Duraturo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
| | - Paola Izzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Marina De Rosa
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
| |
Collapse
|
3
|
Lehtomäki K, Heervä E, Kellokumpu-Lehtinen PL, Mustonen H, Salminen T, Joensuu H, Hermunen K, Boisen MK, Johansen JS, Haglund C, Osterlund P. Transient Changes in Serum CEA, CA19-9, CRP, YKL-40, and IL-6 during Adjuvant Chemotherapy and Survival of Patients with Colorectal Cancer. Int J Mol Sci 2023; 24:ijms24076753. [PMID: 37047727 PMCID: PMC10094778 DOI: 10.3390/ijms24076753] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/22/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023] Open
Abstract
Serum carcinoembryonic antigen (CEA) is frequently monitored to detect colorectal cancer (CRC) recurrence after surgery. The clinical significance of transiently increased CEA during adjuvant chemotherapy is poorly understood. Serum CEA, CA19-9, CRP, YKL-40, and IL-6 were measured before, during, and after adjuvant 5-fluorouracil-based chemotherapy in the randomised LIPSYT study population. The biomarker kinetic patterns were classified into three groups: no increase, a transient increase (≥10% increase followed by a decrease), and a persistent increase during the adjuvant treatment, and the associations of these patterns with disease free-survival (DFS) and overall survival (OS) were investigated by using Cox regression analyses. The findings were validated in two single-centre cohorts that received modern adjuvant chemotherapy. A transient increase in CEA occurred in about a half of the patients during chemotherapy, in all the cohorts. The patients with a transient increase had a roughly similar DFS and OS to the patients with no increase, and a more favourable survival compared to the patients with a persistent increase. In the LIPSYT cohort, the hazard ratio was 0.21 for DFS (CI95% 0.07-0.66) and 0.24 for OS (CI95% 0.08-0.76). Transient increases in CA19-9 and YKL-40 tended to be associated with a favourable survival. A transient increase in CEA during adjuvant chemotherapy is associated with a favourable survival when compared with a persistent increase.
Collapse
Grants
- 2001, 2014, 2018, 2019, 2020, 2021, 2022 Finska Läkaresällskapet
- 2001, 2013, 2019-2020, 2021, 2022-23 Finnish Cancer Foundation
- 2001, 2016, 2017, 2018, 2019, 2020, 2021, 2022, 2023 Competitive State Research Financing of the Expert Responsibility Area of Tampere, Helsinki, and Turku
- Tukisäätiö 2019, 2020, 2022; OOO 2020, 2022 Tampere University Hospital
- 2020-2022 Relander's Foundation
Collapse
Affiliation(s)
- Kaisa Lehtomäki
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland
- Department of Oncology: Tays Cancer Center, Tampere University Hospital, Teiskontie 35, 33520 Tampere, Finland
| | - Eetu Heervä
- Department of Oncology, Turku University Hospital, Hämeentie 11, 20520 Turku, Finland
- Department of Oncology, University of Turku, Kiinanmyllynkatu 10, 20520 Turku, Finland
| | - Pirkko-Liisa Kellokumpu-Lehtinen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland
- Research, Development and Innovation Center, Tampere University Hospital, Teiskontie 35, 33520 Tampere, Finland
| | - Harri Mustonen
- Department of Surgery, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, 00290 Helsinki, Finland
- Research Programs Unit, Translational Cancer Medicine Program, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Tapio Salminen
- Department of Oncology: Tays Cancer Center, Tampere University Hospital, Teiskontie 35, 33520 Tampere, Finland
| | - Heikki Joensuu
- Department of Oncology, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, 00290 Helsinki, Finland
| | - Kethe Hermunen
- Department of Surgery, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, 00290 Helsinki, Finland
| | - Mogens Karsbøl Boisen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls vej 1, DK-2730 Herlev, Denmark
| | - Julia Sidenius Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls vej 1, DK-2730 Herlev, Denmark
- Department of Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital and University of Copenhagen, Borgmester Ib Juuls vej 1, DK-2730 Herlev, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
| | - Caj Haglund
- Department of Surgery, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, 00290 Helsinki, Finland
- Research Programs Unit, Translational Cancer Medicine Program, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Pia Osterlund
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland
- Department of Oncology: Tays Cancer Center, Tampere University Hospital, Teiskontie 35, 33520 Tampere, Finland
- Department of Oncology, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, 00290 Helsinki, Finland
- Department of Gastrointestinal Oncology, Tema Cancer, Karolinska Universitetssjukhuset, Eugeniavägen 3, 17176 Solna, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Solnavägen 1, 17177 Solna, Sweden
| |
Collapse
|
4
|
Hsieh YY, Cheng YW, Wei PL, Yang PM. Repurposing of ingenol mebutate for treating human colorectal cancer by targeting S100 calcium-binding protein A4 (S100A4). Toxicol Appl Pharmacol 2022; 449:116134. [PMID: 35724704 DOI: 10.1016/j.taap.2022.116134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/03/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022]
Abstract
Colorectal cancer (CRC) is the world's second most common cause of cancer-related death. Novel treatments are still urgently needed. S100 calcium-binding protein A4 (S100A4) was demonstrated to be an anticancer therapeutic target. Herein, we found that higher S100A4 expression was associated with a poorer prognosis in publicly available cohorts and a Taiwanese CRC patient cohort. To identify repurposed S100A4 inhibitors, we mined the Connectivity Map (CMap) database for clinical drugs mimicking the S100A4-knockdown gene signature. Ingenol mebutate, derived from the sap of the plant Euphorbia peplus, is approved as a topical treatment for actinic keratosis. The CMap analysis predicted ingenol mebutate as a potent S100A4 inhibitor. Indeed, both messenger RNA and protein levels of S100A4 were attenuated by ingenol mebutate in human CRC cells. In addition, CRC cells with higher S100A4 expressions and/or the wild-type p53 gene were more sensitive to ingenol mebutate, and their migration and invasion were inhibited by ingenol mebutate. Therefore, our results suggest the repurposing of ingenol mebutate for treating CRC by targeting S100A4.
Collapse
Affiliation(s)
- Yao-Yu Hsieh
- Division of Hematology and Oncology, Taipei Medical University Shuang Ho Hospital, New Taipei City 23561, Taiwan; Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Ya-Wen Cheng
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan; Department of R&D, Calgent Biotechnology Co., Ltd., Taipei 10675, Taiwan
| | - Po-Li Wei
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Division of Colorectal Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan; Cancer Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan; Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Pei-Ming Yang
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan.
| |
Collapse
|
5
|
Chen L, Tian B, Liu W, Liang H, You Y, Liu W. Molecular Biomarker of Drug Resistance Developed From Patient-Derived Organoids Predicts Survival of Colorectal Cancer Patients. Front Oncol 2022; 12:855674. [PMID: 35425715 PMCID: PMC9004628 DOI: 10.3389/fonc.2022.855674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 02/10/2022] [Indexed: 12/24/2022] Open
Abstract
The drug 5-fluorouracil (5-Fu) is the critical composition of colorectal cancer (CRC) treatments. Prognostic and predictive molecular biomarkers for CRC patients (CRCpts) treated with 5-Fu-based chemotherapy can provide assistance for tailoring treatment approach. Here, we established a molecular biomarker of 5-Fu resistance derived from colorectal cancer organoids (CRCOs) for predicting the survival of CRCpts. Forty-one CRCO cultures were generated from 50 CRC tumor tissues after surgery (82%). The following experiments revealed a great diversity in drug sensitivity for 10 μM 5-Fu treatment tested by using organoid size change. Fourteen cases (34.1%) were 5-Fu sensitive and the other 27 (65.9%) were resistant. Then, differentially expressed genes (DEGs) associated with 5-Fu resistance were outputted by transcriptome sequencing. In particular, DEGs were generated in two comparison groups: 1) 5-Fu sensitive and resistant untreated CRCOs; 2) CRCOs before 5-Fu treatment and surviving CRCOs after 5-Fu treatment. Some molecules and most of the pathways that have been reported to be involved in 5-Fu resistance were identified in the current research. By using DEGs correlated with 5-Fu resistance and survival of CRCpts, the gene signature and drug-resistant score model (DRSM) containing five molecules were established in The Cancer Genome Atlas (TCGA)-CRC cohort by least absolute shrinkage and selection operator (LASSO) regression analysis and 5-fold cross-validation. Multivariate analysis revealed that drug-resistant score (DRS) was an independent prognostic factor for overall survival (OS) in CRCpts in TCGA-CRC cohort (P < 0.001). Further validation results from four Gene Expression Omnibus (GEO) cohorts elucidated that the DRSM based on five genes related to 5-Fu chemosensitivity and developed from patient-derived organoids can predict survival of CRCpts. Meanwhile, our model could predict the survival of CRCpts in different subgroups. Besides, the difference of molecular pathways, tumor mutational burden (TMB), immune response-related pathways, immune score, stromal score, and immune cell proportion were dissected between DRS-high and DRS-low patients in TCGA-CRC cohort.
Collapse
Affiliation(s)
- Lifeng Chen
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Tian
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wen Liu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haitao Liang
- Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center), Shenzhen, China
| | - Yong You
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weizhen Liu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Hu M, Li C, Wang Z, Ding P, Pei R, Wang Q, Xu H, Xing C. Development of Metal-Organic Framework-Based Dual Antibody Nanoparticles for the Highly Specific Capture and Gradual Release of Circulating Tumor Cells. Front Bioeng Biotechnol 2022; 10:806238. [PMID: 35198544 PMCID: PMC8859420 DOI: 10.3389/fbioe.2022.806238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Circulating tumor cells (CTCs) have been well-established as promising biomarkers that can be leveraged to gauge the prognosis of patients with cancers and to guide patient treatment efforts. Although the scarcity of CTCs within peripheral circulation and the associated phenotypic changes that they exhibit owing to the epithelial-mesenchymal transition (EMT) process make the reliable isolation of these cells very challenging. Recently, several studies have discussed platforms capable of mediating the efficient and sensitive isolation of CTCs, but these approaches are nonetheless subject to certain limitations that preclude their clinical application. For example, these platforms are poorly-suited to minimizing damage in the context of cellular capture and release or the in vitro culture of captured cells for subsequent molecular analyses, which would better enable clinicians to select appropriate precision treatments on an individualized basis. In this study, we report the layer-by-layer assembly approach to synthesize a novel composite nanomaterial consisting of modified zirconium-based metal-organic-frameworks (MOFs) on the surface of magnetic beads with dual antibody surface modifications capable of capturing CTCs without being hampered by the state of cellular EMT process. Our analyses indicated that these dual antibody-modified nanomaterials exhibited greater capture efficiency than that observed for single antibody. Importantly, captured cells can be gradually released following capture and undergo subsequent in vitro proliferation following water molecule-induced MOF structural collapse. This release mechanism, which does not require operator intervention, may be effective as a means of minimizing damage and preserving cellular viability such that cells can be more reliably utilized for downstream molecular analyses and associated treatment planning. To further confirm the potential clinical applicability of the developed nanomaterial, it was successfully utilized for capturing CTCs from peripheral blood samples collected from cases diagnosed with gastrointestinal tumors.
Collapse
Affiliation(s)
- Mingchao Hu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng Li
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China
| | - Zhili Wang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China
| | - Pi Ding
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China
- *Correspondence: Renjun Pei, ; Hua Xu, ; Chungen Xing,
| | | | - Hua Xu
- Department of General Surgery, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
- *Correspondence: Renjun Pei, ; Hua Xu, ; Chungen Xing,
| | - Chungen Xing
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Renjun Pei, ; Hua Xu, ; Chungen Xing,
| |
Collapse
|
7
|
Quiroz TS, Heras FL. Medicina de precisión: rol de la anatomía patológica en diagnóstico y tratamiento personalizado. REVISTA MÉDICA CLÍNICA LAS CONDES 2022. [DOI: 10.1016/j.rmclc.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
8
|
Bonde A, Smith DA, Kikano E, Yoest JM, Tirumani SH, Ramaiya NH. Overview of serum and tissue markers in colorectal cancer: a primer for radiologists. Abdom Radiol (NY) 2021; 46:5521-5535. [PMID: 34415413 DOI: 10.1007/s00261-021-03243-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 08/05/2021] [Accepted: 08/07/2021] [Indexed: 12/17/2022]
Abstract
Serum and tissue tumor markers provide crucial information in the diagnosis, treatment, and follow-up of colorectal cancers. Tissue tumor markers are increasingly used for determination of targeted chemotherapy planning based on genotyping of tumor cells. Recently, plasma-based technique of liquid biopsy is being evaluated for providing tumor biomarkers in the management of colorectal cancer. Tumor markers are commonly used in conjunction with imaging during initial staging, treatment determination, response assessment, and determination of recurrence or metastatic disease. Knowledge of tumor markers and their association with radiological findings is thus crucial for radiologists. Additionally, various novel imaging techniques are being evaluated as potential noninvasive imaging biomarkers to predict tumor genotypes, features, and tumor response. We review and discuss the potential role of these newer imaging techniques.
Collapse
Affiliation(s)
- Apurva Bonde
- Department of Radiology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.
| | - Daniel A Smith
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Elias Kikano
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Jennifer M Yoest
- Department of Pathology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Sree H Tirumani
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Nikhil H Ramaiya
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| |
Collapse
|
9
|
He XS, Zou SY, Yao JL, Yu W, Deng ZY, Wang JR, Gan WJ, Wan S, Yang XQ, Wu H. Transcriptomic Analysis Identifies Complement Component 3 as a Potential Predictive Biomarker for Chemotherapy Resistance in Colorectal Cancer. Front Mol Biosci 2021; 8:763652. [PMID: 34722636 PMCID: PMC8554154 DOI: 10.3389/fmolb.2021.763652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
Objective: 5-fluorouracil- and oxaliplatin-based FOLFOX regimens are mainstay chemotherapeutics for colorectal cancer (CRC) but drug resistance represents a major therapeutic challenge. To improve patient survival, there is a need to identify resistance genes to better understand the mechanisms underlying chemotherapy resistance. Methods: Transcriptomic datasets were retrieved from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases and combined with our own microarray data. Weighted gene co-expression network analysis (WGCNA) was used to dissect the functional networks and hub genes associated with FOLFOX resistance and cancer recurrence. We then conducted analysis of prognosis, profiling of tumor infiltrating immune cells, and pathway overrepresentation analysis to comprehensively elucidate the biological impact of the identified hub gene in CRC. Results: WGCNA analysis identified the complement component 3 (C3) gene as the only hub gene associated with both FOLFOX chemotherapy resistance and CRC recurrence after FOLFOX chemotherapy. Subsequent survival analysis confirmed that high C3 expression confers poor progression-free survival, disease-free survival, and recurrence-free survival. Further correlational analysis revealed significant negative association of C3 expression with sensitivity to oxaliplatin, but not 5-fluorouracil. Moreover, in silico analysis of tumor immune cell infiltration suggested the change of C3 expression could affect tumor microenvironment. Finally, gene set enrichment analysis (GSEA) revealed a hyperactivation of pathways contributing to invasion, metastasis, lymph node spread, and oxaliplatin resistance in CRC samples with C3 overexpression. Conclusion: Our results suggest that high C3 expression is a debilitating factor for FOLFOX chemotherapy, especially for oxaliplatin sensitivity, and C3 may represent a novel biomarker for treatment decision of CRC.
Collapse
Affiliation(s)
- Xiao-Shun He
- Department of Pathology, Medical College of Soochow University and The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Sheng-Yi Zou
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Jia-Lu Yao
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Wangjianfei Yu
- Department of Bioinformatics, Medical College of Soochow University, Soochow University, Suzhou, China
| | - Zhi-Yong Deng
- Department of Pathology, The First People’s Hospital of Kunshan, Kunshan, China
| | - Jing-Ru Wang
- Department of Pathology, Medical College of Soochow University and The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Wen-Juan Gan
- Department of Pathology, Dushu Lake Hospital Affiliated of Soochow University, Soochow University, Suzhou, China
| | - Shan Wan
- Department of Pathology, Medical College of Soochow University and The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Xiao-Qin Yang
- Department of Bioinformatics, Medical College of Soochow University, Soochow University, Suzhou, China
| | - Hua Wu
- Department of Pathology, Medical College of Soochow University and The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
- Department of Pathology, Dushu Lake Hospital Affiliated of Soochow University, Soochow University, Suzhou, China
| |
Collapse
|
10
|
Wang L, Zhao C, Shan H, Jiao Y, Zhang Q, Li X, Yu J, Ding B. Deoxycholic acid-modified microporous SiO 2nanofibers mimicking colorectal microenvironment to optimize radiotherapy-chemotherapy combined therapy. Biomed Mater 2021; 16. [PMID: 34592717 DOI: 10.1088/1748-605x/ac2bbb] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023]
Abstract
Radiotherapy and chemotherapy remain the main therapeutics for colorectal cancer. However, due to their inevitable side effects on nomal tissues, it is necessary to evaluate the toxicity of radio-/chemotherapy regimens. The newly developedin vitrohigh throughput strategy is promising for these assessments. Nevertheless, the currently monolayer culture condition adopted in the preclinical screening processesin vitrohas been proved not so efficient asin vivosince its poor physiological similarity toin vivomicroenvironment. Herein, we fabricated microporous SiO2nanofiber mats and further bioactivated with deoxycholic acid (DCA) to mimic the chemical signals in the colorectal cancer microenvironment forin vitroregimen assessment of radiotherapy and chemotherapy. The colorectal cancer cells contacted with the DCA-modified SiO2nanofiber (SiO2-DCA NF) mats spatially, and the human intestinal epithelial cell on SiO2-DCA NF mats exhibited better x-ray and cisplatin tolerance. The distinguishable irradiation and drug tolerance of cells on SiO2-DCA NF mats indicated that the actual microenvironment of intestine might instruct colorectal cancer differently compared with the common biological experiments. The presented DCA-modified microporous SiO2nanofibrous mats endowing a better mimicry of colorectal micro-environment, would provide a promising platform forin vitroassessment of radio-/chemotherapy regimens.
Collapse
Affiliation(s)
- Lihuan Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Textiles, Donghua University, Shanghai 201620, People's Republic of China.,Guangdong-Hong Kong Joint Laboratory for New Textile Materials, School of Textile Materials and Engineering, Wuyi University, Jiangmen 529020, People's Republic of China
| | - Congzhao Zhao
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, and Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD), Medical College of Soochow University, Suzhou, Jiangsu, 215123, People's Republic of China
| | - Haoru Shan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Textiles, Donghua University, Shanghai 201620, People's Republic of China
| | - Yang Jiao
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, and Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD), Medical College of Soochow University, Suzhou, Jiangsu, 215123, People's Republic of China
| | - Qi Zhang
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, and Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD), Medical College of Soochow University, Suzhou, Jiangsu, 215123, People's Republic of China
| | - Xiaoran Li
- China Innovation Center for Textile Science and Technology, Donghua University, Shanghai 200051, People's Republic of China
| | - Jianyong Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Textiles, Donghua University, Shanghai 201620, People's Republic of China.,China Innovation Center for Textile Science and Technology, Donghua University, Shanghai 200051, People's Republic of China
| | - Bin Ding
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Textiles, Donghua University, Shanghai 201620, People's Republic of China.,China Innovation Center for Textile Science and Technology, Donghua University, Shanghai 200051, People's Republic of China
| |
Collapse
|
11
|
MiR-137 Targets the 3' Untranslated Region of MSH2: Potential Implications in Lynch Syndrome-Related Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13184662. [PMID: 34572889 PMCID: PMC8470766 DOI: 10.3390/cancers13184662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 11/17/2022] Open
Abstract
Mismatch Repair (MMR) gene dysregulation plays a fundamental role in Lynch Syndrome (LS) pathogenesis, a form of hereditary colorectal cancer. Loss or overexpression of key MMR genes leads to genome instability and tumorigenesis; however, the mechanisms controlling MMR gene expression are unknown. One such gene, MSH2, exerts an important role, not only in MMR, but also in cell proliferation, apoptosis, and cell cycle control. In this study, we explored the functions and underlying molecular mechanisms of increased MSH2 expression related to a c.*226A>G variant in the 3'untranslated (UTR) region of MSH2 that had been previously identified in a subject clinically suspected of LS. Bioinformatics identified a putative binding site for miR-137 in this region. To verify miRNA targeting specificity, we performed luciferase gene reporter assays using a MSH2 3'UTR psiCHECK-2 vector in human SW480 cells over-expressing miR-137, which showed a drastic reduction in luciferase activity (p > 0.0001). This effect was abolished by site-directed mutagenesis of the putative miR-137 seed site. Moreover, in these cells we observed that miR-137 levels were inversely correlated with MSH2 expression levels. These results were confirmed by results in normal and tumoral tissues from the patient carrying the 3'UTR c.*226A>G variant in MSH2. Finally, miR-137 overexpression in SW480 cells significantly suppressed cell proliferation in a time- and dose-dependent manner (p < 0.0001), supporting a role for MSH2 in apoptosis and cell proliferation processes. Our findings suggest miR-137 helps control MSH2 expression via its 3'UTR and that dysregulation of this mechanism appears to promote tumorigenesis in colon cells.
Collapse
|
12
|
Liu HY, Zhang HS, Liu MY, Li HM, Wang XY, Wang M. GLS1 depletion inhibited colorectal cancer proliferation and migration via redox/Nrf2/autophagy-dependent pathway. Arch Biochem Biophys 2021; 708:108964. [PMID: 34119480 DOI: 10.1016/j.abb.2021.108964] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/23/2021] [Accepted: 06/07/2021] [Indexed: 01/17/2023]
Abstract
Cancer cells can metabolize glutamine to replenish TCA cycle intermediates for cell survival. Glutaminase (GLS1) is over-expressed in multiple cancers, including colorectal cancer (CRC). However, the role of GLS1 in colorectal cancer development has not yet fully elucidated. In this study, we found that GLS1 levels were significantly increased in CRC cells. Knockdown of GLS1 by shRNAs as well as GLS1 inhibitor BPTES decreased DLD1 and SW480 cell proliferation, colony formation and migration. Knockdown of GLS1 as well as BPTES induced reactive oxygen species (ROS) production, down-regulation of GSH/GSSG ratio, an decrease in Nrf2 protein expression and an increase in cytoplasmic Nrf2 protein expression in DLD1 and SW480 cells. Furthermore, Knockdown of GLS1 as well as BPTES inhibited autophagy pathway, antioxidant NAC and Nrf2 activator could reversed inhibition of GLS1-mediated an decrease in autophagic flux in DLD1 and SW480 cells. Depletion of GLS1-induced inhibition of DLD1 and SW480 CRC cell proliferation, colony formation and migration was reversed by autophagy inducer rapamycin. These results suggest that targeting GLS1 might be a new potential therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Hui-Yun Liu
- Faculty of Environment and Life, Beijing University of Technology, Pingleyuan 100#, District of Chaoyang, Beijing, 100124, China
| | - Hong-Sheng Zhang
- Faculty of Environment and Life, Beijing University of Technology, Pingleyuan 100#, District of Chaoyang, Beijing, 100124, China.
| | - Min-Yao Liu
- Faculty of Environment and Life, Beijing University of Technology, Pingleyuan 100#, District of Chaoyang, Beijing, 100124, China
| | - Hong-Ming Li
- Faculty of Environment and Life, Beijing University of Technology, Pingleyuan 100#, District of Chaoyang, Beijing, 100124, China
| | - Xin-Yu Wang
- Faculty of Environment and Life, Beijing University of Technology, Pingleyuan 100#, District of Chaoyang, Beijing, 100124, China
| | - Miao Wang
- Faculty of Environment and Life, Beijing University of Technology, Pingleyuan 100#, District of Chaoyang, Beijing, 100124, China
| |
Collapse
|
13
|
Lead Time and Prognostic Role of Serum CEA, CA19-9, IL-6, CRP, and YKL-40 after Adjuvant Chemotherapy in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13153892. [PMID: 34359796 PMCID: PMC8345682 DOI: 10.3390/cancers13153892] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 12/24/2022] Open
Abstract
In colorectal cancer (CRC), 20-50% of patients relapse after curative-intent surgery with or without adjuvant therapy. We investigated the lead times and prognostic value of post-adjuvant (8 months from randomisation to adjuvant treatment) serum CEA, CA19-9, IL-6, CRP, and YKL-40. We included 147 radically resected stage II-IV CRC treated with 24 weeks of adjuvant 5-fluorouracil-based chemotherapy in the phase III LIPSYT-study (ISRCTN98405441). All 147 were included in lead time analysis, but 12 relapsing during adjuvant therapy were excluded from post-adjuvant analysis. Elevated post-adjuvant CEA, IL-6, and CRP were associated with impaired disease-free survival (DFS) with hazard ratio (HR) 5.21 (95% confidence interval 2.32-11.69); 3.72 (1.99-6.95); 2.58 (1.18-5.61), respectively, and elevated IL-6 and CRP with impaired overall survival (OS) HR 3.06 (1.64-5.73); 3.41 (1.55-7.49), respectively. Elevated post-adjuvant IL-6 in CEA-normal patients identified a subgroup with impaired DFS. HR 3.12 (1.38-7.04) and OS, HR 3.20 (1.39-7.37). The lead times between the elevated biomarker and radiological relapse were 7.8 months for CEA and 10.0-53.1 months for CA19-9, IL-6, CRP, and YKL-40, and the lead time for the five combined was 27.3 months. Elevated post-adjuvant CEA, IL-6, and CRP were associated with impaired DFS. The lead time was shortest for CEA.
Collapse
|
14
|
Potential of olive oil and its phenolic compounds as therapeutic intervention against colorectal cancer: a comprehensive review. Br J Nutr 2021; 128:1257-1273. [PMID: 34338174 DOI: 10.1017/s0007114521002919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Colorectal cancer (CRC) is one of the major causes of death across the world and incidence rate of CRC increasing alarmingly each passing year. Diet, genomic anomalies, inflammation and deregulated signalling pathways are among the major causes of CRC. Because of numerous side effects of CRC therapies available now, researchers all over the world looking for alternative treatment/preventive strategy with lesser/no side effects. Olive oil which is part of Mediterranean diet contains numerous phenolic compounds that fight against free radicals and inflammation and also well-known for protective role against CRC. The current review focused on the recent evidences where olive oil and its phenolic compounds such as hydroxytyrosol, oleuropein and oleocanthal showed activities against CRC as well to analyse the cellular and molecular signalling mechanism through which these compounds act on. These compounds shown to combat CRC by reducing proliferation, migration, invasion and angiogenesis through regulation of numerous signalling pathways including MAPK pathway, PI3K-Akt pathway and Wnt/β-catenin pathway and at the same time, induce apoptosis in different CRC model. However, further research is an absolute necessity to establish these compounds as nutritional supplements and develop therapeutic strategy in CRC.
Collapse
|
15
|
Turano M, Cammarota F, Duraturo F, Izzo P, De Rosa M. A Potential Role of IL-6/IL-6R in the Development and Management of Colon Cancer. MEMBRANES 2021; 11:membranes11050312. [PMID: 33923292 PMCID: PMC8145725 DOI: 10.3390/membranes11050312] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 01/13/2023]
Abstract
Colorectal cancer (CRC) is the third most frequent cancer worldwide and the second greatest cause of cancer deaths. About 75% of all CRCs are sporadic cancers and arise following somatic mutations, while about 10% are hereditary cancers caused by germline mutations in specific genes. Several factors, such as growth factors, cytokines, and genetic or epigenetic alterations in specific oncogenes or tumor-suppressor genes, play a role during the adenoma-carcinoma sequence. Recent studies have reported an increase in interleukin-6 (IL-6) and soluble interleukin-6 receptor (sIL-6R) levels in the sera of patients affected by colon cancer that correlate with the tumor size, suggesting a potential role for IL-6 in colon cancer progression. IL-6 is a pleiotropic cytokine showing both pro- and anti-inflammatory roles. Two different types of IL-6 signaling are known. Classic IL-6 signaling involves the binding of IL-6 to its membrane receptor on the surfaces of target cells; alternatively, IL-6 binds to sIL-6R in a process called IL-6 trans-signaling. The activation of IL-6 trans-signaling by metalloproteinases has been described during colon cancer progression and metastasis, involving a shift from membrane-bound interleukin-6 receptor (IL-6R) expression on the tumor cell surface toward the release of soluble IL-6R. In this review, we aim to shed light on the role of IL-6 signaling pathway alterations in sporadic colorectal cancer and the development of familial polyposis syndrome. Furthermore, we evaluate the possible roles of IL-6 and IL-6R as biomarkers useful in disease follow-up and as potential targets for therapy, such as monoclonal antibodies against IL-6 or IL-6R, or a food-based approach against IL-6.
Collapse
Affiliation(s)
- Mimmo Turano
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy;
| | - Francesca Cammarota
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.C.); (F.D.); (P.I.)
- Ceinge Biotecnologie Avanzate, 80131 Naples, Italy
| | - Francesca Duraturo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.C.); (F.D.); (P.I.)
- Ceinge Biotecnologie Avanzate, 80131 Naples, Italy
| | - Paola Izzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.C.); (F.D.); (P.I.)
- Ceinge Biotecnologie Avanzate, 80131 Naples, Italy
| | - Marina De Rosa
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.C.); (F.D.); (P.I.)
- Ceinge Biotecnologie Avanzate, 80131 Naples, Italy
- Correspondence:
| |
Collapse
|
16
|
Silva VSE, Abdallah EA, de Brito ABC, Braun AC, Tariki MS, de Mello CAL, Calsavara VF, Riechelmann R, Chinen LTD. Baseline and Kinetic Circulating Tumor Cell Counts Are Prognostic Factors in a Prospective Study of Metastatic Colorectal Cancer. Diagnostics (Basel) 2021; 11:diagnostics11030502. [PMID: 33809053 PMCID: PMC7999095 DOI: 10.3390/diagnostics11030502] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/01/2021] [Accepted: 03/07/2021] [Indexed: 12/24/2022] Open
Abstract
The discovery of predictive biomarkers in metastatic colorectal cancer (mCRC) is essential to improve clinical outcomes. Recent data suggest a potential role of circulating tumor cells (CTCs) as prognostic indicators. We conducted a follow-on analysis from a prospective study of consecutive patients with mCRC. CTC analysis was conducted at two timepoints: baseline (CTC1; before starting chemotherapy), and two months after starting treatment (CTC2). CTC isolation/quantification were completed by ISET® (Rarecells, France). CTC expressions of drug resistance-associated proteins were evaluated. Progression-free survival (PFS) and overall survival (OS) were estimated by the Kaplan–Meier method. Seventy-five patients were enrolled from May 2012 to May 2014. A CTC1 cut-off of >1.5 CTCs/mL was associated with an inferior median OS compared to lower values. A difference of CTC2−CTC1 > 5.5 CTCs/mL was associated with a reduced median PFS. By multivariate analysis, CTC1 > 1.5 CTCs/mL was an independent prognostic factor for worse OS. Multi-drug resistance protein-1 (MRP-1) expression was associated with poor median OS. CTC baseline counts, kinetics, and MRP-1 expression were predictive of clinical outcomes. Larger studies are warranted to explore the potential clinical benefit of treating mCRC patients with targeted therapeutic regimens guided by CTC findings.
Collapse
Affiliation(s)
- Virgílio Souza e Silva
- Department of Medical Oncology, A.C. Camargo Cancer Center, São Paulo 01509-900, Brazil; (V.S.eS.); (A.B.C.d.B.); (M.S.T.); (C.A.L.d.M.); (R.R.)
| | - Emne Ali Abdallah
- International Research Center, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (E.A.A.); (A.C.B.); (V.F.C.)
| | - Angelo Borsarelli Carvalho de Brito
- Department of Medical Oncology, A.C. Camargo Cancer Center, São Paulo 01509-900, Brazil; (V.S.eS.); (A.B.C.d.B.); (M.S.T.); (C.A.L.d.M.); (R.R.)
| | - Alexcia Camila Braun
- International Research Center, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (E.A.A.); (A.C.B.); (V.F.C.)
| | - Milena Shizue Tariki
- Department of Medical Oncology, A.C. Camargo Cancer Center, São Paulo 01509-900, Brazil; (V.S.eS.); (A.B.C.d.B.); (M.S.T.); (C.A.L.d.M.); (R.R.)
| | - Celso Abdon Lopes de Mello
- Department of Medical Oncology, A.C. Camargo Cancer Center, São Paulo 01509-900, Brazil; (V.S.eS.); (A.B.C.d.B.); (M.S.T.); (C.A.L.d.M.); (R.R.)
| | - Vinicius Fernando Calsavara
- International Research Center, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (E.A.A.); (A.C.B.); (V.F.C.)
| | - Rachel Riechelmann
- Department of Medical Oncology, A.C. Camargo Cancer Center, São Paulo 01509-900, Brazil; (V.S.eS.); (A.B.C.d.B.); (M.S.T.); (C.A.L.d.M.); (R.R.)
| | - Ludmilla Thomé Domingos Chinen
- International Research Center, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (E.A.A.); (A.C.B.); (V.F.C.)
- Correspondence:
| |
Collapse
|
17
|
Abstract
In this Special Issue of Cancers, the latest insights on biomarkers in cancers are presented in 33 up-to-the-minute research papers and reviews summing up the tremendous progress in this interesting and important field of research. The recent development of new therapeutic approaches has provided clinicians with more efficient tools than ever before for the treatment of cancerous diseases. However, choosing the right option requires to [...].
Collapse
|
18
|
Ferretti E, Carlomagno S, Pesce S, Muccio L, Obino V, Greppi M, Solari A, Setti C, Marcenaro E, Della Chiesa M, Sivori S. Role of the Main Non HLA-Specific Activating NK Receptors in Pancreatic, Colorectal and Gastric Tumors Surveillance. Cancers (Basel) 2020; 12:E3705. [PMID: 33321719 PMCID: PMC7763095 DOI: 10.3390/cancers12123705] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022] Open
Abstract
Human NK cells can control tumor growth and metastatic spread thanks to their powerful cytolytic activity which relies on the expression of an array of activating receptors. Natural cytotoxicity receptors (NCRs) NKG2D and DNAM-1 are those non-HLA-specific activating NK receptors that are mainly involved in sensing tumor transformation by the recognition of different ligands, often stress-induced molecules, on the surface of cancer cells. Tumors display several mechanisms aimed at dampening/evading NK-mediated responses, a relevant fraction of which is based on the downregulation of the expression of activating receptors and/or their ligands. In this review, we summarize the role of the main non-HLA-specific activating NK receptors, NCRs, NKG2D and DNAM-1, in controlling tumor growth and metastatic spread in solid malignancies affecting the gastrointestinal tract with high incidence in the world population, i.e., pancreatic ductal adenocarcinoma (PDAC), colorectal cancer (CRC), and gastric cancer (GC), also describing the phenotypic and functional alterations induced on NK cells by their tumor microenvironment.
Collapse
Affiliation(s)
- Elisa Ferretti
- Centro di Eccellenza per la Ricerca Biomedica, University of Genoa, 16132 Genoa, Italy;
| | - Simona Carlomagno
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Silvia Pesce
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Letizia Muccio
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Valentina Obino
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Marco Greppi
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Agnese Solari
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Chiara Setti
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Emanuela Marcenaro
- Dipartimento di Medicina Sperimentale (DIMES) and Centro di Eccellenza per la Ricerca Biomedica, University of Genoa, 16132 Genoa, Italy;
| | - Mariella Della Chiesa
- Dipartimento di Medicina Sperimentale (DIMES) and Centro di Eccellenza per la Ricerca Biomedica, University of Genoa, 16132 Genoa, Italy;
| | - Simona Sivori
- Dipartimento di Medicina Sperimentale (DIMES) and Centro di Eccellenza per la Ricerca Biomedica, University of Genoa, 16132 Genoa, Italy;
| |
Collapse
|
19
|
Wang N, He JX, Jia GZ, Wang K, Zhou S, Wu T, He XL. The lncRNA XIST promotes colorectal cancer cell growth through regulating the miR-497-5p/FOXK1 axis. Cancer Cell Int 2020; 20:553. [PMID: 33298041 PMCID: PMC7727145 DOI: 10.1186/s12935-020-01647-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/09/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Recent studies suggest that long noncoding RNAs (lncRNAs) play an important role in tumorigenesis. As a newly identified lncRNA, the role of XIST in colorectal cancer (CRC) has not been established. Here, we sought to characterize the role of XIST and its associated regulatory network in CRC cells. METHODS Expression of XIST mRNA, miR-497-5p, and forkhead box k1 (FOXK1) in CRC cells and tissues were detected using quantitative real-time polymerase chain reaction (qRT-PCR). Proliferation and apoptosis of CRC cells were determined using the CCK-8 cell counting assay and flow cytometry. The rate of cell migration and invasion was determined using a transwell assay. The relationships between XIST, miR-497-5p, and FOXK1 were predicted and confirmed using a dual-luciferase reporter assay. Expression of FOXK1 protein was quantified by Western blot. RESULTS XIST and FOXK1 expression were significantly upregulated in CRC tissues and cell lines, while miR-497-5p expression was downregulated. XIST knockdown significantly suppressed CRC cell proliferation, migration, and invasion. Silencing of XIST also reversed the downregulation of miR-497-5p and upregulation of FOXK1. Moreover, blocking XIST expression was shown to inhibit CRC tumor growth in vivo and the effects were antagonized by the loss of miR-497-5p. miR-497-5p was shown to act as a sponge of XIST and also targeted FOXK1 in CRC cells. CONCLUSIONS XIST was shown to promote the malignancy of CRC cells by competitively binding to miR-497-5p, resulting in an increase in FOXK1 expression. These results suggest that targeting of XIST may represent a possible treatment for CRC.
Collapse
Affiliation(s)
- Nan Wang
- Department of General Surgery, Tangdu Hospital, the Air Force Medical University, 710038, Xi'an, China
| | - Jia-Xing He
- Department of General Surgery, Tangdu Hospital, the Air Force Medical University, 710038, Xi'an, China
| | - Guo-Zhan Jia
- Department of General Surgery, Tangdu Hospital, the Air Force Medical University, 710038, Xi'an, China
| | - Ke Wang
- Department of General Surgery, Tangdu Hospital, the Air Force Medical University, 710038, Xi'an, China
| | - Shuai Zhou
- Department of General Surgery, Tangdu Hospital, the Air Force Medical University, 710038, Xi'an, China
| | - Tao Wu
- Department of General Surgery, Tangdu Hospital, the Air Force Medical University, 710038, Xi'an, China.
| | - Xian-Li He
- Department of General Surgery, Tangdu Hospital, the Air Force Medical University, 710038, Xi'an, China.
| |
Collapse
|
20
|
Resectability and Ablatability Criteria for the Treatment of Liver Only Colorectal Metastases: Multidisciplinary Consensus Document from the COLLISION Trial Group. Cancers (Basel) 2020; 12:cancers12071779. [PMID: 32635230 PMCID: PMC7407587 DOI: 10.3390/cancers12071779] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/24/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023] Open
Abstract
The guidelines for metastatic colorectal cancer crudely state that the best local treatment should be selected from a ‘toolbox’ of techniques according to patient- and treatment-related factors. We created an interdisciplinary, consensus-based algorithm with specific resectability and ablatability criteria for the treatment of colorectal liver metastases (CRLM). To pursue consensus, members of the multidisciplinary COLLISION and COLDFIRE trial expert panel employed the RAND appropriateness method (RAM). Statements regarding patient, disease, tumor and treatment characteristics were categorized as appropriate, equipoise or inappropriate. Patients with ECOG≤2, ASA≤3 and Charlson comorbidity index ≤8 should be considered fit for curative-intent local therapy. When easily resectable and/or ablatable (stage IVa), (neo)adjuvant systemic therapy is not indicated. When requiring major hepatectomy (stage IVb), neo-adjuvant systemic therapy is appropriate for early metachronous disease and to reduce procedural risk. To downstage patients (stage IVc), downsizing induction systemic therapy and/or future remnant augmentation is advised. Disease can only be deemed permanently unsuitable for local therapy if downstaging failed (stage IVd). Liver resection remains the gold standard. Thermal ablation is reserved for unresectable CRLM, deep-seated resectable CRLM and can be considered when patients are in poor health. Irreversible electroporation and stereotactic body radiotherapy can be considered for unresectable perihilar and perivascular CRLM 0-5cm. This consensus document provides per-patient and per-tumor resectability and ablatability criteria for the treatment of CRLM. These criteria are intended to aid tumor board discussions, improve consistency when designing prospective trials and advance intersociety communications. Areas where consensus is lacking warrant future comparative studies.
Collapse
|
21
|
Saridemir S, Güven HE, Aksel B, Doğan L. Serum AMDL DR-70 levels: a new concept in the diagnosis and follow-up of colorectal carcinoma. Biomark Med 2020; 14:621-628. [PMID: 32608998 DOI: 10.2217/bmm-2020-0004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Aim: The purpose of this study was to determine the diagnostic potential of DR-70 assay in patients with colorectal cancer and to investigate the relationship between serum DR-70 levels and the biological characteristic of the tumor. Patients & methods: The experimental group included patients who were diagnosed with colorectal adenocarcinoma after biopsy specimen. The control group of this study consisted of patients whose total colonoscopy was reported as normal. Results: Serum levels of DR-70 was found to be significantly higher in patients with colorectal cancer than healthy participants (p = 0.001). Receiver operating curve analyses indicated a cut-off value of 1.69 μg/ml for serum DR-70 levels. Stage of the disease, grade of the tumor, number of metastatic lymph nodes and microsatellite instability status were significantly related to serum DR-70 levels (p = 0.001, p = 0.001, p = 0.001 and p = 0.002, respectively). Conclusion: It can be concluded that serum levels of DR-70 can be regarded as an indicator for the diagnosis of colorectal cancer.
Collapse
Affiliation(s)
- Serdar Saridemir
- Department of General Surgery, Ankara Oncology Training & Research Hospital, Health Sciences University, Ankara, Turkey
| | - Hikmet E Güven
- Department of General Surgery, Ankara Gülhane Training & Research Hospital, Health Sciences University, Ankara, Turkey
| | - Bülent Aksel
- Department of General Surgery, Ankara Oncology Training & Research Hospital, Health Sciences University, Ankara, Turkey
| | - Lütfi Doğan
- Department of General Surgery, Ankara Oncology Training & Research Hospital, Health Sciences University, Ankara, Turkey
| |
Collapse
|
22
|
Damilakis E, Mavroudis D, Sfakianaki M, Souglakos J. Immunotherapy in Metastatic Colorectal Cancer: Could the Latest Developments Hold the Key to Improving Patient Survival? Cancers (Basel) 2020; 12:E889. [PMID: 32268531 PMCID: PMC7225960 DOI: 10.3390/cancers12040889] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy has considerably increased the number of anticancer agents in many tumor types including metastatic colorectal cancer (mCRC). Anti-PD-1 (programmed death 1) and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) immune checkpoint inhibitors (ICI) have been shown to benefit the mCRC patients with mismatch repair deficiency (dMMR) or high microsatellite instability (MSI-H). However, ICI is not effective in mismatch repair proficient (pMMR) colorectal tumors, which constitute a large population of patients. Several clinical trials evaluating the efficacy of immunotherapy combined with chemotherapy, radiation therapy, or other agents are currently ongoing to extend the benefit of immunotherapy to pMMR mCRC cases. In dMMR patients, MSI testing through immunohistochemistry and/or polymerase chain reaction can be used to identify patients that will benefit from immunotherapy. Next-generation sequencing has the ability to detect MSI-H using a low amount of nucleic acids and its application in clinical practice is currently being explored. Preliminary data suggest that radiomics is capable of discriminating MSI from microsatellite stable mCRC and may play a role as an imaging biomarker in the future. Tumor mutational burden, neoantigen burden, tumor-infiltrating lymphocytes, immunoscore, and gastrointestinal microbiome are promising biomarkers that require further investigation and validation.
Collapse
Affiliation(s)
- Emmanouil Damilakis
- Department of Medical Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece; (D.M.); (J.S.)
| | - Dimitrios Mavroudis
- Department of Medical Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece; (D.M.); (J.S.)
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - Maria Sfakianaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - John Souglakos
- Department of Medical Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece; (D.M.); (J.S.)
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| |
Collapse
|