1
|
Montero-Calle A, Garranzo-Asensio M, Moreno-Casbas MT, Campuzano S, Barderas R. Autoantibodies in cancer: a systematic review of their clinical role in the most prevalent cancers. Front Immunol 2024; 15:1455602. [PMID: 39234247 PMCID: PMC11371560 DOI: 10.3389/fimmu.2024.1455602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
Although blood autoantibodies were initially associated with autoimmune diseases, multiple evidence have been accumulated showing their presence in many types of cancer. This has opened their use in clinics, since cancer autoantibodies might be useful for early detection, prognosis, and monitoring of cancer patients. In this review, we discuss the different techniques available for their discovery and validation. Additionally, we discuss here in detail those autoantibody panels verified in at least two different reports that should be more likely to be specific of each of the four most incident cancers. We also report the recent developed kits for breast and lung cancer detection mostly based on autoantibodies and the identification of novel therapeutic targets because of the screening of the cancer humoral immune response. Finally, we discuss unsolved issues that still need to be addressed for the implementation of cancer autoantibodies in clinical routine for cancer diagnosis, prognosis, and/or monitoring.
Collapse
Affiliation(s)
- Ana Montero-Calle
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Maria Teresa Moreno-Casbas
- Investén-isciii, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Research Center Network for Frailty and Healthy Ageing (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Susana Campuzano
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | - Rodrigo Barderas
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Research Center Network for Frailty and Healthy Ageing (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
2
|
Qiu C, Wang X, Batson SA, Wang B, Casiano CA, Francia G, Zhang JY. A Luminex Approach to Develop an Anti-Tumor-Associated Antigen Autoantibody Panel for the Detection of Prostate Cancer in Racially/Ethnically Diverse Populations. Cancers (Basel) 2023; 15:4064. [PMID: 37627091 PMCID: PMC10452333 DOI: 10.3390/cancers15164064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
(1) Background: Autoantibodies to tumor-associated antigens (TAAs) have emerged as promising cancer biomarkers. Luminex technology offers a powerful approach for the simultaneous detection of multiple anti-TAA autoantibodies. (2) Methods: We aimed to utilize Luminex technology to evaluate and optimize a panel of anti-TAAs autoantibodies for detecting prostate cancer (PCa), which included autoantibodies to fourteen TAAs. A total of 163 serum samples (91 PCa, 72 normal controls) were screened to determine the levels of the autoantibodies using the Luminex assay. (3) Results: Twelve autoantibodies exhibited significantly high frequencies ranging from 19.8% to 51.6% in the PCa group. Receiver operating characteristic (ROC) curve analysis revealed area under the curve (AUC) values ranging from 0.609 to 0.868 for the twelve autoantibodies individually. We further confirmed the performance of the HSP60 autoantibody by using an enzyme-linked immunosorbent assay (ELISA) in a larger sample comprising 200 PCa sera, 20 benign prostatic hyperplasia (BPH) sera, and 137 normal control sera. The results obtained from the Luminex assay were consistent with the ELISA findings. We developed a panel consisting of three autoantibodies (p16, IMP2, and HSP60) which achieved an impressive AUC of 0.910 with a sensitivity of 71.4% and a specificity of 95.8%. The panel was also evaluated in PCa patients from different races/ethnicities with the best performance observed in distinguishing the Hispanic American patients with PCa from normal controls. (4) Conclusions: We developed an anti-TAA autoantibody panel for the detection of PCa that exhibits promising performance. This panel holds significant potential as a high-throughput tool to facilitate PCa detection.
Collapse
Affiliation(s)
- Cuipeng Qiu
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (C.Q.); (X.W.); (S.A.B.); (B.W.)
| | - Xiao Wang
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (C.Q.); (X.W.); (S.A.B.); (B.W.)
| | - Serina A. Batson
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (C.Q.); (X.W.); (S.A.B.); (B.W.)
| | - Bofei Wang
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (C.Q.); (X.W.); (S.A.B.); (B.W.)
| | - Carlos A. Casiano
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA;
| | - Giulio Francia
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (C.Q.); (X.W.); (S.A.B.); (B.W.)
| | - Jian-Ying Zhang
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (C.Q.); (X.W.); (S.A.B.); (B.W.)
| |
Collapse
|
3
|
Ke B, Liang ZK, Li B, Wang XJ, Liu N, Liang H, Zhang RP. EDIL3 is a potential prognostic biomarker that correlates with immune infiltrates in gastric cancer. PeerJ 2023; 11:e15559. [PMID: 37576496 PMCID: PMC10422953 DOI: 10.7717/peerj.15559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 05/24/2023] [Indexed: 08/15/2023] Open
Abstract
Background EDIL3, which contains epidermal growth factor-like repeats and discoidin I-like domains, is a secretory protein that plays an important role in embryonic development and various illnesses. However, the biological function of EDIL3 in gastric cancer (GC) is still unclear. The objective of this research was to explore the role and potential mechanism of EDIL3 in GC. Methods In this study, we used the GEPIA, HPA, MethSurv, SMART, STRING, GeneMANIA, LinkedOmics TIMER, TIMER2.0, TISIDB, and RNAactDrug databases to comprehensively analyze the roles of EDIL3 in GC. To validate the in silico findings, EDIL3 expression was measured in our collected GC tissues. Meanwhile, several in vitro experiments were performed to test the function of EDIL3 in GC. Results We found that EDIL3 was highly expressed in GC and associated with adverse clinical features. In vitro assays revealed that EDIL3 promoted the proliferation, migration, and invasion of GC cells. The functions of EDIL3 and co-expression genes were significantly associated with extracellular structure organization and matrix receptor interaction. EDIL3 expression was positively associated with numerous tumor-infiltrating immune cells and their biomarkers. Conclusion This study determined that EDIL3 may function as an oncogene and is associated with immune infiltration in GC. EDIL3 could be used as a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Bin Ke
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zheng-Kai Liang
- Department of Gastrointestinal Surgery, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Bin Li
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xue-Jun Wang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Ning Liu
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Han Liang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Ru-Peng Zhang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
4
|
Alorda-Clara M, Torrens-Mas M, Morla-Barcelo PM, Martinez-Bernabe T, Sastre-Serra J, Roca P, Pons DG, Oliver J, Reyes J. Use of Omics Technologies for the Detection of Colorectal Cancer Biomarkers. Cancers (Basel) 2022; 14:817. [PMID: 35159084 PMCID: PMC8834235 DOI: 10.3390/cancers14030817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most frequently diagnosed cancers with high mortality rates, especially when detected at later stages. Early detection of CRC can substantially raise the 5-year survival rate of patients, and different efforts are being put into developing enhanced CRC screening programs. Currently, the faecal immunochemical test with a follow-up colonoscopy is being implemented for CRC screening. However, there is still a medical need to describe biomarkers that help with CRC detection and monitor CRC patients. The use of omics techniques holds promise to detect new biomarkers for CRC. In this review, we discuss the use of omics in different types of samples, including breath, urine, stool, blood, bowel lavage fluid, or tumour tissue, and highlight some of the biomarkers that have been recently described with omics data. Finally, we also review the use of extracellular vesicles as an improved and promising instrument for biomarker detection.
Collapse
Affiliation(s)
- Marina Alorda-Clara
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (T.M.-B.); (J.S.-S.); (P.R.); (D.G.P.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Edificio S, E-07120 Palma de Mallorca, Illes Balears, Spain
| | - Margalida Torrens-Mas
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (T.M.-B.); (J.S.-S.); (P.R.); (D.G.P.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Edificio S, E-07120 Palma de Mallorca, Illes Balears, Spain
- Translational Research in Aging and Longevity (TRIAL) Group, Instituto de Investigación Sanitaria Illes Balears (IdISBa), E-07120 Palma de Mallorca, Illes Balears, Spain
| | - Pere Miquel Morla-Barcelo
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (T.M.-B.); (J.S.-S.); (P.R.); (D.G.P.)
| | - Toni Martinez-Bernabe
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (T.M.-B.); (J.S.-S.); (P.R.); (D.G.P.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Edificio S, E-07120 Palma de Mallorca, Illes Balears, Spain
| | - Jorge Sastre-Serra
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (T.M.-B.); (J.S.-S.); (P.R.); (D.G.P.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Edificio S, E-07120 Palma de Mallorca, Illes Balears, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03) Instituto Salud Carlos III, E-28029 Madrid, Madrid, Spain
| | - Pilar Roca
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (T.M.-B.); (J.S.-S.); (P.R.); (D.G.P.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Edificio S, E-07120 Palma de Mallorca, Illes Balears, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03) Instituto Salud Carlos III, E-28029 Madrid, Madrid, Spain
| | - Daniel Gabriel Pons
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (T.M.-B.); (J.S.-S.); (P.R.); (D.G.P.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Edificio S, E-07120 Palma de Mallorca, Illes Balears, Spain
| | - Jordi Oliver
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (T.M.-B.); (J.S.-S.); (P.R.); (D.G.P.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Edificio S, E-07120 Palma de Mallorca, Illes Balears, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03) Instituto Salud Carlos III, E-28029 Madrid, Madrid, Spain
| | - Jose Reyes
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (T.M.-B.); (J.S.-S.); (P.R.); (D.G.P.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Edificio S, E-07120 Palma de Mallorca, Illes Balears, Spain
- Servicio Aparato Digestivo, Hospital Comarcal de Inca, E-07300 Inca, Illes Balears, Spain
| |
Collapse
|
5
|
Proteomic Analyses of Fibroblast- and Serum-Derived Exosomes Identify QSOX1 as a Marker for Non-invasive Detection of Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13061351. [PMID: 33802764 PMCID: PMC8002505 DOI: 10.3390/cancers13061351] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/12/2021] [Accepted: 03/13/2021] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Early diagnosis of colorectal cancer (CRC) is crucial to improve patient outcomes. The tumour microenvironment immediately adapts to malignant transformations, including the activation of fibroblasts in the connective tissue nearby. In this study, we investigated fibroblast activity-related protein secretion via extracellular vesicles (EVs). QSOX1, a protein identified to be significantly reduced in activated fibroblasts and derived EVs, was also found to be significantly reduced in circulating blood plasma EVs of CRC patients as compared to control patients. Hence, blood plasma EV-associated QSOX1 represents a promising platform for diagnostic CRC screening. Abstract The treatment of colorectal cancer (CRC) has improved during the last decades, but methods for crucial early diagnosis are yet to be developed. The influence of the tumour microenvironment on liquid biopsies for early cancer diagnostics are gaining growing interest, especially with emphasis on exosomes (EXO), a subgroup of extracellular vesicles (EVs). In this study, we established paired cancer-associated (CAFs) and normal fibroblasts (NF) from 13 CRC patients and investigated activation status-related protein abundance in derived EXOs. Immunohistochemical staining of matched patient tissue was performed and an independent test cohort of CRC patient plasma-derived EXOs was assessed by ELISA. A total of 11 differentially abundant EV proteins were identified between NFs and CAFs. In plasma EXOs, the CAF-EXO enriched protein EDIL3 was elevated, while the NF-EXO enriched protein QSOX1 was diminished compared to whole plasma. Both markers were significantly reduced in patient-matched CRC tissue compared to healthy colon tissue. In an independent test cohort, a significantly reduced protein abundance of QSOX1 was observed in plasma EXOs from CRC patients compared to controls and diagnostic ROC curve analysis revealed an AUC of 0.904. In conclusion, EXO-associated QSOX1 is a promising novel marker for early diagnosis and non-invasive risk stratification in CRC.
Collapse
|
6
|
Ganig N, Baenke F, Thepkaysone ML, Lin K, Rao VS, Wong FC, Polster H, Schneider M, Helm D, Pecqueux M, Seifert AM, Seifert L, Weitz J, Rahbari NN, Kahlert C. Proteomic Analyses of Fibroblast- and Serum-Derived Exosomes Identify QSOX1 as a Marker for Non-invasive Detection of Colorectal Cancer. Cancers (Basel) 2021. [PMID: 33802764 DOI: 10.3390/cancers130613510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Abstract
The treatment of colorectal cancer (CRC) has improved during the last decades, but methods for crucial early diagnosis are yet to be developed. The influence of the tumour microenvironment on liquid biopsies for early cancer diagnostics are gaining growing interest, especially with emphasis on exosomes (EXO), a subgroup of extracellular vesicles (EVs). In this study, we established paired cancer-associated (CAFs) and normal fibroblasts (NF) from 13 CRC patients and investigated activation status-related protein abundance in derived EXOs. Immunohistochemical staining of matched patient tissue was performed and an independent test cohort of CRC patient plasma-derived EXOs was assessed by ELISA. A total of 11 differentially abundant EV proteins were identified between NFs and CAFs. In plasma EXOs, the CAF-EXO enriched protein EDIL3 was elevated, while the NF-EXO enriched protein QSOX1 was diminished compared to whole plasma. Both markers were significantly reduced in patient-matched CRC tissue compared to healthy colon tissue. In an independent test cohort, a significantly reduced protein abundance of QSOX1 was observed in plasma EXOs from CRC patients compared to controls and diagnostic ROC curve analysis revealed an AUC of 0.904. In conclusion, EXO-associated QSOX1 is a promising novel marker for early diagnosis and non-invasive risk stratification in CRC.
Collapse
Affiliation(s)
- Nicole Ganig
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Franziska Baenke
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - May-Linn Thepkaysone
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Kuailu Lin
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Venkatesh S Rao
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Fang Cheng Wong
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Heike Polster
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Martin Schneider
- MS-based Protein Analysis Unit, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Dominic Helm
- MS-based Protein Analysis Unit, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Mathieu Pecqueux
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Adrian M Seifert
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, D-69120 Heidelberg, Germany
| | - Lena Seifert
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, D-69120 Heidelberg, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Nuh N Rahbari
- Department of Surgery, University Medicine Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, D-68167 Mannheim, Germany
| | - Christoph Kahlert
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, D-69120 Heidelberg, Germany
| |
Collapse
|
7
|
Gallardo-Gómez M, De Chiara L, Álvarez-Chaver P, Cubiella J. Colorectal cancer screening and diagnosis: omics-based technologies for development of a non-invasive blood-based method. Expert Rev Anticancer Ther 2021; 21:723-738. [PMID: 33507120 DOI: 10.1080/14737140.2021.1882858] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Colorectal cancer (CRC) is one of the most important health problems in the Western world. In order to reduce the burden of the disease, two strategies are proposed: screening and prompt detection in symptomatic patients. Although diagnosis and prevention are mainly based on colonoscopy, fecal hemoglobin detection has been widely implemented as a noninvasive strategy. Various studies aiming to discover blood-based biomarkers have recently emerged.Areas covered: The burgeoning omics field provides diverse high-throughput approaches for CRC blood-based biomarker discovery. In this review, we appraise the most robust and commonly used technologies within the fields of genomics, transcriptomics, epigenomics, proteomics, and metabolomics, together with their targeted validation approaches. We summarize the transference process from the discovery phase until clinical translation. Finally, we review the best candidate biomarkers and their potential clinical applicability.Expert opinion: Some available biomarkers are promising, especially in the field of epigenomics: DNA methylation and microRNA. Transference requires the joint collaboration of basic researchers, intellectual property experts, technology transfer officers and clinicians. Blood-based biomarkers will be selected not only based on their diagnostic accuracy and cost but also on their reliability, applicability to clinical analysis laboratories and their acceptance by the population.
Collapse
Affiliation(s)
- María Gallardo-Gómez
- Department of Biochemistry, Genetics and Immunology, University of Vigo, Vigo, Spain.,Biomedical Research Center (CINBIO), University of Vigo, Vigo, Spain
| | - Loretta De Chiara
- Department of Biochemistry, Genetics and Immunology, University of Vigo, Vigo, Spain.,Biomedical Research Center (CINBIO), University of Vigo, Vigo, Spain
| | - Paula Álvarez-Chaver
- Proteomics Unit, Service of Structural Determination, Proteomics and Genomics, Center for Scientific and Technological Research Support (CACTI), University of Vigo, Vigo, Spain
| | - Joaquin Cubiella
- Department of Gastroenterology, Hospital Universitario De Ourense, Ourense, Spain.,Instituto De Investigación Sanitaria Galicia Sur, Ourense, Spain.,Centro De Investigación Biomédica En Red Enfermedades Hepáticas Y Digestivas, Ourense, Spain
| |
Collapse
|
8
|
Chen X, Sun J, Wang X, Yuan Y, Cai L, Xie Y, Fan Z, Liu K, Jiao X. A Meta-Analysis of Proteomic Blood Markers of Colorectal Cancer. Curr Med Chem 2021; 28:1176-1196. [PMID: 32338203 DOI: 10.2174/0929867327666200427094054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/23/2020] [Accepted: 03/24/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Early diagnosis will significantly improve the survival rate of colorectal cancer (CRC); however, the existing methods for CRC screening were either invasive or inefficient. There is an emergency need for novel markers in CRC's early diagnosis. Serum proteomics has gained great potential in discovering novel markers, providing markers that reflect the early stage of cancer and prognosis prediction of CRC. In this paper, the results of proteomics of CRC studies were summarized through a meta-analysis in order to obtain the diagnostic efficiency of novel markers. METHODS A systematic search on bibliographic databases was performed to collect the studies that explore blood-based markers for CRC applying proteomics. The detection and validation methods, as well as the specificity and sensitivity of the biomarkers in these studies, were evaluated. Newcastle- Ottawa Scale (NOS) case-control studies version was used for quality assessment of included studies. RESULTS Thirty-four studies were selected from 751 studies, in which markers detected by proteomics were summarized. In total, fifty-nine proteins were classified according to their biological function. The sensitivity, specificity, or AUC varied among these markers. Among them, Mammalian STE20-like protein kinase 1/ Serine threonine kinase 4 (MST1/STK4), S100 calcium-binding protein A9 (S100A9), and Tissue inhibitor of metalloproteinases 1 (TIMP1) were suitable for effect sizes merging, and their diagnostic efficiencies were recalculated after merging. MST1/STK4 obtained a sensitivity of 68% and a specificity of 78%. S100A9 achieved a sensitivity of 72%, a specificity of 83%, and an AUC of 0.88. TIMP1 obtained a sensitivity of 42%, a specificity of 88%, and an AUC of 0.71. CONCLUSION MST1/STK4, S100A9, and TIMP1 showed excellent performance for CRC detection. Several other markers also presented optimized diagnostic efficacy for CRC early detection, but further verification is still needed before they are suitable for clinical use. The discovering of more efficient markers will benefit CRC treatment.
Collapse
Affiliation(s)
- Xiang Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jiayu Sun
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Xue Wang
- Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Yumeng Yuan
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Leshan Cai
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Yanxuan Xie
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Zhiqiang Fan
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Kaixi Liu
- Shantou Central Hospital, Shantou, Guangdong 515041, China
| | - Xiaoyang Jiao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, China
| |
Collapse
|
9
|
Yang SH, Liu CT, Hong CQ, Huang ZY, Wang HZ, Wei LF, Lin YW, Guo HP, Peng YH, Xu YW. Autoantibodies against p53, MMP-7, and Hsp70 as Potential Biomarkers for Detection of Nonmelanoma Skin Cancers. DISEASE MARKERS 2021; 2021:5592693. [PMID: 34336006 PMCID: PMC8289574 DOI: 10.1155/2021/5592693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/31/2021] [Indexed: 02/05/2023]
Abstract
Basal cell carcinoma (BCC) and squamous cell carcinoma (SCC) are two predominant histological types of nonmelanoma skin cancer (NMSC), lacking effective early diagnostic markers. In this study, we assessed the diagnostic value of autoantibodies against p53, MMP-7, and Hsp70 in skin SCC and BCC. ELISA was performed to detect levels of autoantibodies in sera from 101 NMSC patients and 102 normal controls, who were recruited from the Cancer Hospital of Shantou University Medical College. A receiver operator characteristic curve was used to evaluate the diagnostic value. The serum levels of autoantibodies against p53, MMP-7, and Hsp70 were higher in NMSCs than those in the normal controls (all P < 0.01). The AUC of the three-autoantibody panel was 0.841 (95% CI: 0.788-0.894) with the sensitivity and specificity of 60.40% and 91.20% when differentiating NMSCs from normal controls. Furthermore, measurement of this panel could differentiate early-stage skin cancer patients from normal controls (AUC: 0.851; 95% CI: 0.793-0.908). Data from Oncomine showed that the level of p53 mRNA was elevated in BCC (P < 0.05), and the Hsp70 mRNA was upregulated in SCC (P < 0.001). This serum three-autoantibody panel might function in assisting the early diagnosis of NMSC.
Collapse
Affiliation(s)
- Shi-Han Yang
- Department of Dermatology and Venereology, Affiliated Shantou Hospital of Sun Yat-sen University, 114 Waima Road, Shantou 515041, China
| | - Can-Tong Liu
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou 515041, China
- Precision Medicine Research Center, Shantou University Medical College, 22 Xinling Road, Shantou 515041, China
| | - Chao-Qun Hong
- Department of Oncological Laboratory Research, The Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou 515041, China
| | - Ze-Yuan Huang
- Department of Dermatology and Venereology, Affiliated Shantou Hospital of Sun Yat-sen University, 114 Waima Road, Shantou 515041, China
| | - Huan-Zhu Wang
- Department of Dermatology and Venereology, Affiliated Shantou Hospital of Sun Yat-sen University, 114 Waima Road, Shantou 515041, China
| | - Lai-Feng Wei
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou 515041, China
- Precision Medicine Research Center, Shantou University Medical College, 22 Xinling Road, Shantou 515041, China
| | - Yi-Wei Lin
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou 515041, China
- Precision Medicine Research Center, Shantou University Medical College, 22 Xinling Road, Shantou 515041, China
| | - Hai-Peng Guo
- Department of Head and Neck Surgery, The Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou 515041, China
| | - Yu-Hui Peng
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou 515041, China
- Precision Medicine Research Center, Shantou University Medical College, 22 Xinling Road, Shantou 515041, China
- Guangdong Esophageal Cancer Research Institute, Shantou University Medical College, 22 Xinling Road, Shantou 515041, China
| | - Yi-Wei Xu
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou 515041, China
- Precision Medicine Research Center, Shantou University Medical College, 22 Xinling Road, Shantou 515041, China
- Guangdong Esophageal Cancer Research Institute, Shantou University Medical College, 22 Xinling Road, Shantou 515041, China
| |
Collapse
|
10
|
Ruan Z, Deng H, Liang M, Xu Z, Lai M, Ren H, Deng X, Su X. Overexpression of long non-coding RNA00355 enhances proliferation, chemotaxis, and metastasis in colon cancer via promoting GTF2B-mediated ITGA2. Transl Oncol 2020; 14:100947. [PMID: 33227664 PMCID: PMC7689553 DOI: 10.1016/j.tranon.2020.100947] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/24/2022] Open
Abstract
LncRNA LINC00355 promotes colon cancer malignancy. LncRNA LINC00355 positively regulates ITGA2 via recruiting GTF2B. LncRNA LINC00355 positively regulates GTF2B-mediated ITGA2 to promote colon cancer. This study proposes a novel targeted strategy for cancer treatment.
Long non-coding RNAs (LncRNAs) can regulate physiological and pathological functions, exhibiting a wide range of roles in cell biology. Moreover, many lncRNAs are dysregulated in various cancers, including colon cancer. In this study, we investigated the role of the lncRNA LINC00355 in colon cancer, after first establishing its interaction with GTF2B, and ITGA2 on the LncMap database. The predicted relationships between the lncRNA LINC00355, GTF2B, and ITGA2 were identified using luciferase reporter assay, RIP, and ChIP experiments. Western blot analysis and RT-qPCR were applied to determine expression pattern of lncRNA LINC00355 and ITGA2 in colon cancer cells. Additionally, EdU, TUNEL, Cell-adhesion and Transwell assay was used for the detection of the effects of this axis on proliferation, apoptosis, adhesion, chemotaxis and metastasis. LncRNA LINC00355 targeted IGFBP2 through the recruitment of GTF2B. LncRNA LINC00355 was highly expressed in colon cancer cells, and overexpression of lncRNA LINC00355 increased the expression of IGFBP2 and GTF2B, and thereby promoted the proliferation, chemotaxis, invasion, and migration in colon cancer. In summary, downregulation of lncRNA LINC00355 in colon cancer inhibited tumor growth in colon cancer through effects on the GTF2B/IGFBP2 axis.
Collapse
Affiliation(s)
- Zhiyan Ruan
- School of Pharmacy, Guangdong Food & Drug Vocational College, No. 321, Longdong North Road, Tianhe District, Guangzhou 510520, Guangdong Province, PR China
| | - Hongling Deng
- School of Pharmacy, Guangdong Food & Drug Vocational College, No. 321, Longdong North Road, Tianhe District, Guangzhou 510520, Guangdong Province, PR China
| | - Minhua Liang
- School of Pharmacy, Guangdong Food & Drug Vocational College, No. 321, Longdong North Road, Tianhe District, Guangzhou 510520, Guangdong Province, PR China
| | - Zhe Xu
- School of Pharmacy, Guangdong Food & Drug Vocational College, No. 321, Longdong North Road, Tianhe District, Guangzhou 510520, Guangdong Province, PR China
| | - Manxiang Lai
- School of Pharmacy, Guangdong Food & Drug Vocational College, No. 321, Longdong North Road, Tianhe District, Guangzhou 510520, Guangdong Province, PR China
| | - Hong Ren
- School of Pharmacy, Guangdong Food & Drug Vocational College, No. 321, Longdong North Road, Tianhe District, Guangzhou 510520, Guangdong Province, PR China
| | - Xiangliang Deng
- School of Chinese Medicine, Guangdong Pharmaceutical University, No. 280, East Ring Road, Guangzhou University Town, Guangzhou 510006, Guangdong Province, PR China.
| | - Xinguo Su
- School of Pharmacy, Guangdong Food & Drug Vocational College, No. 321, Longdong North Road, Tianhe District, Guangzhou 510520, Guangdong Province, PR China.
| |
Collapse
|
11
|
Honma N, Inoue T, Tsuchiya N, Koizumi A, Yamamoto R, Nara T, Kanda S, Huang M, Numakura K, Saito M, Narita S, Satoh S, Habuchi T. Prognostic value of plasminogen activator inhibitor-1 in biomarker exploration using multiplex immunoassay in patients with metastatic renal cell carcinoma treated with axitinib. Health Sci Rep 2020; 3:e197. [PMID: 33088925 PMCID: PMC7559632 DOI: 10.1002/hsr2.197] [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: 04/13/2020] [Revised: 08/24/2020] [Accepted: 09/17/2020] [Indexed: 11/08/2022] Open
Abstract
Background and Aims Vascular endothelial growth factor‐directed therapies play a significant role in patients with metastatic renal cell carcinoma (mRCC). Biomarkers for predicting treatment efficacy and resistance are required to develop personalized medicine. We evaluated multiple serum cytokine levels in patients with mRCC treated with axitinib to explore predictive biomarkers. Methods From September 2012 to October 2015, serum samples were collected from 44 patients with mRCC before treatment and 4 weeks after axitinib initiation. Bio‐Plex Pro Human Cancer Biomarker Panels 1 and 2 were used to measure levels of 34 serum biomarkers related to angiogenesis and cell proliferation. Results Patients with partial response or stable disease had significantly decreased serum plasminogen activator inhibitor‐1 (PAI‐1) level from pre‐treatment to 4 weeks after axitinib initiation compared with those with progressive disease (P = .022). The median progression‐free survival (PFS) and median overall survival (OS) in patients with increased serum PAI‐1 level from pre‐treatment to 4 weeks after axitinib initiation were significantly shorter than those with decreased serum PAI‐1 level (P = .027 and P = .026, respectively). Increased serum PAI‐1 level from pre‐treatment to 4 weeks after axitinib initiation was an independent prognostic marker for shorter PFS and OS in multivariate analyses (P = .015 and P = .032, respectively). The immunohistochemical staining intensity of PAI‐1 in tumor specimens was significantly associated with Fuhrman grade and presence of distant metastasis (P = .026 and P = .010, respectively). Conclusions The initial change in serum PAI‐1 level in the early stage of axitinib treatment could be a useful prognostic biomarker in patients with mRCC.
Collapse
Affiliation(s)
- Naoko Honma
- Department of Urology Akita University Graduate School of Medicine Akita Japan
| | - Takamitsu Inoue
- Department of Urology Akita University Graduate School of Medicine Akita Japan.,AMED-CREST Japan Science and Technology Agency Tokyo Japan
| | - Norihiko Tsuchiya
- Department of Urology Akita University Graduate School of Medicine Akita Japan.,Department of Urology Yamagata University Faculty of Medicine Yamagata Japan
| | - Atsushi Koizumi
- Department of Urology Akita University Graduate School of Medicine Akita Japan
| | - Ryohei Yamamoto
- Department of Urology Akita University Graduate School of Medicine Akita Japan
| | - Taketoshi Nara
- Department of Urology Akita University Graduate School of Medicine Akita Japan
| | - Sohei Kanda
- Department of Urology Akita University Graduate School of Medicine Akita Japan
| | - Mingguo Huang
- Department of Urology Akita University Graduate School of Medicine Akita Japan
| | - Kazuyuki Numakura
- Department of Urology Akita University Graduate School of Medicine Akita Japan
| | - Mitsuru Saito
- Department of Urology Akita University Graduate School of Medicine Akita Japan
| | - Shintaro Narita
- Department of Urology Akita University Graduate School of Medicine Akita Japan.,AMED-CREST Japan Science and Technology Agency Tokyo Japan
| | - Shigeru Satoh
- Center for Kidney Disease and Transplantation Akita University Hospital Akita Japan
| | - Tomonori Habuchi
- Department of Urology Akita University Graduate School of Medicine Akita Japan.,AMED-CREST Japan Science and Technology Agency Tokyo Japan
| |
Collapse
|
12
|
Gan X, Wang T, Chen ZY, Zhang KH. Blood-derived molecular signatures as biomarker panels for the early detection of colorectal cancer. Mol Biol Rep 2020; 47:8159-8168. [DOI: 10.1007/s11033-020-05838-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022]
|
13
|
High expression of PLAC1 in colon cancer as a predictor of poor prognosis: A study based on TCGA data. Gene 2020; 763:145072. [PMID: 32827679 DOI: 10.1016/j.gene.2020.145072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/25/2020] [Accepted: 08/17/2020] [Indexed: 01/19/2023]
Abstract
Colon cancer is one of the most common diseases in the world with both a high incidence and high mortality. PLAC1 is activated and expressed in many cancers. We aim to explore the relationship between PLAC1 expression and prognosis in colon cancer patient. The RNA-Seq expression data and clinical information of colon cancer were downloaded from The Cancer Genome Atlas (TCGA) database. Differentially expressed PLAC1 was obtained by the Wilcoxon signed-rank test; the significance difference being that PLAC1 was more highly expressed in tumor rather than normal tissue (p < 0.01). Then patients were classified into high and low risk groups by different risk scores, and the Kaplan-Meier survival analysis showed that colon cancer patients with a high PLAC1 expression had a poorer prognosis than low PLAC1 expression patients (p = 0.0031). Next, in analyzing the clinical pathology associated with PLAC1 expression, logistic regression showed that PLAC1 was expressed high in stage (OR = 4.11 for I vs. IV), lymph nodes (OR = 1.73 for N0 vs. N1+), distant metastasis (OR = 2.8 for M0 vs. M1), and status (OR = 22.81 for normal vs. tumor). Univariate and multivariate cox analyses were employed to identify that PLAC1 could be regarded as an independent prognostic factor. Univariate cox analysis showed PLAC1 had a correlation to overall survival (OS) (HR: 0.46, 95% CI: 0.28-0.77, p = 0.003). Multivariate cox analysis revealed that PLAC1 (HR: 0.51, 95% CI: 0.30-0.86, p = 0.012) could be regarded as an Independent prognostic factor. We also used quantitative real-time polymerase chain reaction (qRT-PCR) to test if PLAC1 was differently expressed in cell lines. The qRT-PCR obtained the significant results that PLAC1 expressed high in colon cancer cell lines (p < 0.05). Finally, Gene Set Enrichment Analysis (GSEA) was utilized to show 14 enriched signaling pathways. Our study discovered that high expression of PLAC1 predicts poor prognosis in colon cancer patients, providing a new biomarker, which can assist physicians in finding new diagnostic and therapy methods for colon cancer.
Collapse
|
14
|
Quinchia J, Echeverri D, Cruz-Pacheco AF, Maldonado ME, Orozco J. Electrochemical Biosensors for Determination of Colorectal Tumor Biomarkers. MICROMACHINES 2020; 11:E411. [PMID: 32295170 PMCID: PMC7231317 DOI: 10.3390/mi11040411] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 12/15/2022]
Abstract
The accurate determination of specific tumor markers associated with cancer with non-invasive or minimally invasive procedures is the most promising approach to improve the long-term survival of cancer patients and fight against the high incidence and mortality of this disease. Quantification of biomarkers at different stages of the disease can lead to an appropriate and instantaneous therapeutic action. In this context, the determination of biomarkers by electrochemical biosensors is at the forefront of cancer diagnosis research because of their unique features such as their versatility, fast response, accurate quantification, and amenability for multiplexing and miniaturization. In this review, after briefly discussing the relevant aspects and current challenges in the determination of colorectal tumor markers, it will critically summarize the development of electrochemical biosensors to date to this aim, highlighting the enormous potential of these devices to be incorporated into the clinical practice. Finally, it will focus on the remaining challenges and opportunities to bring electrochemical biosensors to the point-of-care testing.
Collapse
Affiliation(s)
- Jennifer Quinchia
- Max Planck Tandem Group in Nanobioengineering, University of Antioquia, Complejo Ruta N, Calle 67 No. 52-20, Medellín 050010, Colombia; (J.Q.); (D.E.); (A.F.C.-P.)
| | - Danilo Echeverri
- Max Planck Tandem Group in Nanobioengineering, University of Antioquia, Complejo Ruta N, Calle 67 No. 52-20, Medellín 050010, Colombia; (J.Q.); (D.E.); (A.F.C.-P.)
| | - Andrés Felipe Cruz-Pacheco
- Max Planck Tandem Group in Nanobioengineering, University of Antioquia, Complejo Ruta N, Calle 67 No. 52-20, Medellín 050010, Colombia; (J.Q.); (D.E.); (A.F.C.-P.)
| | - María Elena Maldonado
- Grupo Impacto de los Componentes Alimentarios en la Salud, School of Dietetics and Human Nutrition, University of Antioquia, A.A. 1226, Medellín 050010, Colombia;
| | - Jahir Orozco
- Max Planck Tandem Group in Nanobioengineering, University of Antioquia, Complejo Ruta N, Calle 67 No. 52-20, Medellín 050010, Colombia; (J.Q.); (D.E.); (A.F.C.-P.)
| |
Collapse
|
15
|
Garranzo-Asensio M, Guzmán-Aránguez A, Povedano E, Ruiz-Valdepeñas Montiel V, Poves C, Fernandez-Aceñero MJ, Montero-Calle A, Solís-Fernández G, Fernandez-Diez S, Camps J, Arenas M, Rodríguez-Tomàs E, Joven J, Sanchez-Martinez M, Rodriguez N, Dominguez G, Yáñez-Sedeño P, Pingarrón JM, Campuzano S, Barderas R. Multiplexed monitoring of a novel autoantibody diagnostic signature of colorectal cancer using HaloTag technology-based electrochemical immunosensing platform. Theranostics 2020; 10:3022-3034. [PMID: 32194852 PMCID: PMC7053203 DOI: 10.7150/thno.42507] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 01/02/2020] [Indexed: 12/15/2022] Open
Abstract
Background and Purpose: The humoral immune response in cancer patients can be used for early detection of the disease. Autoantibodies raised against tumor-associated antigens (TAAs) are promising clinical biomarkers for reliable cancer diagnosis, prognosis, and therapy monitoring. In this study, an electrochemical disposable multiplexed immunosensing platform able to integrate difficult- and easy-to-express colorectal cancer (CRC) TAAs is reported for the sensitive determination of eight CRC-specific autoantibodies. Methods: The electrochemical immunosensing approach involves the use of magnetic microcarriers (MBs) as solid supports modified with covalently immobilized HaloTag fusion proteins for the selective capture of specific autoantibodies. After magnetic capture of the modified MBs onto screen-printed carbon working electrodes, the amperometric responses measured using the hydroquinone (HQ)/H2O2 system were related to the levels of autoantibodies in plasma. Results: The biosensing platform was applied to the analysis of autoantibodies against 8 TAAs described for the first time in this work in plasma samples from healthy asymptomatic individuals (n=3), and patients with high-risk of developing CRC (n=3), and from patients already diagnosed with colorectal (n=3), lung (n=2) or breast (n=2) cancer. The developed bioplatform demonstrated an improved discrimination between CRC patients and controls (asymptomatic healthy individuals and breast and lung cancer patients) compared to an ELISA-like luminescence test. Conclusions: The proposed methodology uses a just-in-time produced protein in a simpler protocol, with low sample volume, and involves cost-effective instrumentation, which could be used in a high-throughput manner for reliable population screening to facilitate the detection of early CRC patients at affordable cost.
Collapse
Affiliation(s)
- María Garranzo-Asensio
- Departamento de Bioquímica y Biología Molecular, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, 28037 Madrid, Spain
- UFIEC, Chronic Disease Programme, Instituto de Salud Carlos III, Majadahonda 28220, Madrid, Spain
| | - Ana Guzmán-Aránguez
- Departamento de Bioquímica y Biología Molecular, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Eloy Povedano
- Departamento de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Víctor Ruiz-Valdepeñas Montiel
- Departamento de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Carmen Poves
- Gastroenterology Unit, Hospital Universitario Clínico San Carlos, E-28040, Madrid, Spain
| | | | - Ana Montero-Calle
- UFIEC, Chronic Disease Programme, Instituto de Salud Carlos III, Majadahonda 28220, Madrid, Spain
| | | | | | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d´Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus (Spain)
| | - Meritxell Arenas
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d´Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus (Spain)
| | - Elisabeth Rodríguez-Tomàs
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d´Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus (Spain)
- Department of Radiation Oncology, Hospital Universitari Sant Joan, Institut d´Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus (Spain)
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d´Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus (Spain)
| | | | - Nuria Rodriguez
- Medical Oncology Department, Hospital Universitario La Paz, E-28046, Madrid, Spain
| | - Gemma Dominguez
- Departamento de Medicina, Facultad de Medicina, Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, E-28029, Madrid, Spain
| | - Paloma Yáñez-Sedeño
- Departamento de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - José Manuel Pingarrón
- Departamento de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Susana Campuzano
- Departamento de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Rodrigo Barderas
- UFIEC, Chronic Disease Programme, Instituto de Salud Carlos III, Majadahonda 28220, Madrid, Spain
| |
Collapse
|
16
|
Wang H, Li X, Zhou D, Huang J. Autoantibodies as biomarkers for colorectal cancer: A systematic review, meta-analysis, and bioinformatics analysis. Int J Biol Markers 2019; 34:334-347. [PMID: 31588830 DOI: 10.1177/1724600819880906] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Colorectal cancer is a very common cancer worldwide. Serum tumor-associated autoantibodies (TAAbs), especially the anti-p53 autoantibody, may be promising biomarkers to detect early-stage colorectal cancer. This study aimed to identify all known autoantibodies and their value in colorectal cancer diagnosis, as well as exploring the underlying connections and mechanisms through a bioinformatics analysis. Databases were used to select available articles of TAAbs in colorectal cancer. In a meta-analysis of the anti-p53 autoantibody, the diagnostic odds ratio and area under the curve (AUC) of the summary receiver-operating characteristic (SROC) curve were calculated using Stata 12.0 and Meta-Disc 1.4. We identified 73 articles including 199 single autoantibodies and 42 multiple autoantibodies. The maximum value of Youden's index was 0.76, combining c-MYC, p53, cyclin B1, p62, Koc, IMP1, and survivin. The diagnostic odds ratio for anti-p53 autoantibody at all stages was 10.86 (95% CI 8.40, 14.06) with low heterogeneity (I2 = 40.3%) and the AUC of the SROC curve was 0.82. For the anti-p53 autoantibody in early-stage colorectal cancer, the diagnostic odds ratio was 4.82 (95% CI 2.95, 7.87) with heterogeneity (I2 = 7.9%) and the AUC of the SROC curve was 0.72. Eighty-seven autoantibodies were selected for bioinformatics analyses. We found that the most enriched functional terms and protein-protein interactions may relate to the mechanism of autoantibody generation. In summary, our study summarized the diagnostic value of TAAbs in colorectal cancer, either as single molecules or in combination. Bioinformatics analyses may be a new approach to explore the mechanism of autoantibody generation.
Collapse
Affiliation(s)
- Hejing Wang
- Department of Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Xiaojin Li
- Department of Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Donghu Zhou
- Department of Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Jian Huang
- Department of Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| |
Collapse
|
17
|
Marín‐Vicente C, Mendes M, los Ríos V, Fernández‐Aceñero MJ, Casal JI. Identification and Validation of Stage‐Associated Serum Biomarkers in Colorectal Cancer Using MS‐Based Procedures. Proteomics Clin Appl 2019; 14:e1900052. [PMID: 31502404 DOI: 10.1002/prca.201900052] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 09/03/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Consuelo Marín‐Vicente
- Department of Molecular BiomedicineCentro de Investigaciones Biológicas (CIB‐CSIC) Madrid Spain
- Proteomics facilityCentro de Investigaciones Biológicas (CIB‐CSIC) Madrid Spain
| | - Marta Mendes
- Department of Molecular BiomedicineCentro de Investigaciones Biológicas (CIB‐CSIC) Madrid Spain
| | - Vivian los Ríos
- Proteomics facilityCentro de Investigaciones Biológicas (CIB‐CSIC) Madrid Spain
| | | | - J. Ignacio Casal
- Department of Molecular BiomedicineCentro de Investigaciones Biológicas (CIB‐CSIC) Madrid Spain
| |
Collapse
|
18
|
O'Halloran C, McCulloch L, Rentoul L, Alexander J, Hope JC, Gunn-Moore DA. Cytokine and Chemokine Concentrations as Biomarkers of Feline Mycobacteriosis. Sci Rep 2018; 8:17314. [PMID: 30470763 PMCID: PMC6251861 DOI: 10.1038/s41598-018-35571-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/02/2018] [Indexed: 12/16/2022] Open
Abstract
Mycobacteriosis is an emerging zoonotic disease of domestic cats and timely, accurate diagnosis is currently challenging. To identify differential cytokine/chemokine concentrations in serum/plasma of cats, which could be diagnostic biomarkers of infection we analysed plasma/serum from 116 mycobacteria-infected cats, 16 healthy controls and six cats hospitalised for unrelated reasons was analysed using the Milliplex MAP Feline Cytokine Magnetic Bead multiplex assay. Three cytokines; sFAS, IL-13 and IL-4 were reduced while seven; GM-CSF, IL-2, PDGF-BB, IL-8, KC, RANTES and TNF-α were elevated in mycobacteria-infected cats compared to healthy controls. However, IL-8 and KC concentrations were not significantly different from cats hospitalised for other reasons. Elevations in TNF-α and PDGF-BB may have potential to identify M. bovis and M. microti infected cats specifically while GM-CSF, IL-2 and FLT3L were increased in MTBC infected cats. This study demonstrates potential use of feline tuberculosis as a spontaneously occurring model of this significant human disease. Cytokine profiling has clear diagnostic potential for mycobacteriosis of cats and could be used discriminate tuberculous from non-tuberculous disease to rapidly inform on zoonotic risk. Future work should focus on the in-field utility of these findings to establish diagnostic sensitivity and specificity of these markers.
Collapse
Affiliation(s)
- C O'Halloran
- Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, Scotland, EH25 9RG, UK. conor.o'
| | - L McCulloch
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, Scotland, EH16 4SB, UK
| | - L Rentoul
- MilliporeSigma (a Division of Merck KGaA, Darmstadt, Germany), 3050, Spruce Street, St. Louis, MO, USA
| | - J Alexander
- Waltham Centre for Pet Nutrition, Leicestershire, UK
| | - J C Hope
- Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, Scotland, EH25 9RG, UK
| | - D A Gunn-Moore
- Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, Scotland, EH25 9RG, UK
| |
Collapse
|
19
|
Parsa SF, Vafajoo A, Rostami A, Salarian R, Rabiee M, Rabiee N, Rabiee G, Tahriri M, Yadegari A, Vashaee D, Tayebi L, Hamblin MR. Early diagnosis of disease using microbead array technology: A review. Anal Chim Acta 2018; 1032:1-17. [PMID: 30143206 PMCID: PMC6152944 DOI: 10.1016/j.aca.2018.05.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 12/31/2022]
Abstract
Early diagnosis of diseases (before they become advanced and incurable) is essential to reduce morbidity and mortality rates. With the advent of novel technologies in clinical laboratory diagnosis, microbead-based arrays have come to be recognized as an efficient approach, that demonstrates useful advantages over traditional assay methods for multiple disease-related biomarkers. Multiplexed microbead assays provide a robust, rapid, specific, and cost-effective approach for high-throughput and simultaneous screening of many different targets. Biomolecular binding interactions occur after applying a biological sample (such as blood plasma, saliva, cerebrospinal fluid etc.) containing the target analyte(s) to a set of microbeads with different ligand-specificities that have been coded in planar or suspension arrays. The ligand-receptor binding activity is tracked by optical signals generated by means of flow cytometry analysis in the case of suspension arrays, or by image processing devices in the case of planar arrays. In this review paper, we discuss diagnosis of cancer, neurological and infectious diseases by using optically-encoded microbead-based arrays (both multiplexed and single-analyte assays) as a reliable tool for detection and quantification of various analytes.
Collapse
Affiliation(s)
- Sanam Foroutan Parsa
- Biomaterials Group, Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Atieh Vafajoo
- Biomaterials Group, Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Azin Rostami
- Biomaterials Group, Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Reza Salarian
- Biomedical Engineering Department, Maziar University, Noor, Royan, Iran
| | - Mohammad Rabiee
- Biomaterials Group, Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Navid Rabiee
- Department of Chemistry, Shahid Beheshti University, Tehran, Iran
| | - Ghazal Rabiee
- Department of Chemistry, Shahid Beheshti University, Tehran, Iran
| | | | - Amir Yadegari
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - Daryoosh Vashaee
- Electrical and Computer Engineering Department, North Carolina State University, Raleigh, NC 27606, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA; Biomaterials and Advanced Drug Delivery Laboratory, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
20
|
Zhang H, Wang Z, Ma R, Wu J, Feng J. MicroRNAs as biomarkers for the progression and prognosis of colon carcinoma. Int J Mol Med 2018; 42:2080-2088. [PMID: 30066832 PMCID: PMC6108873 DOI: 10.3892/ijmm.2018.3792] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/10/2018] [Indexed: 12/14/2022] Open
Abstract
Early detection is critical for the treatment of colon carcinoma. However, current biomarkers for its diagnosis and prognosis are insufficient and improvement is required. Aberrantly expressed microRNAs (miRNAs/miRs) in colon carcinoma have been identified to function as potential diagnostic and prognostic biomarkers. In the present study, 245 differentially expressed miRNAs between colon carcinoma and normal tissues were identified by a bioinformatics analysis of a dataset from The Cancer Genome Atlas. A six-miRNA (miR-149, miR-3189, miR-3677, miR-3917, miR-4999 and miR-6854) prognostic prediction system was established, which is able to independently and effectively predict the prognosis of colon carcinoma patients [P<0.001, area under the receiver operating characteristic curve (AUC)=0.763]. Furthermore, the six miRNAs were highly correlated with the tumor-nodes-metastasis (TNM) stage and were able to distinguish between different stages (high vs. low TNM stage, P<0.001). Of note, combination of the six-miRNA signature and TNM stage provides an improved prediction of the patient's prognosis (AUC=0.797). Functional enrichment analysis revealed the possible mechanistic involvement of these predictive miRNAs in cancer-associated biological processes and pathways. Taken together, the present study demonstrated the promising potential of the novel six-miRNA model as an independent factor for the prediction of the progression and prognosis of colon carcinoma.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210000, P.R. China
| | - Zhuo Wang
- Laboratory of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210000, P.R. China
| | - Rong Ma
- Laboratory of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210000, P.R. China
| | - Jianzhong Wu
- Laboratory of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210000, P.R. China
| | - Jifeng Feng
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210000, P.R. China
| |
Collapse
|
21
|
Lee PY, Chin SF, Low TY, Jamal R. Probing the colorectal cancer proteome for biomarkers: Current status and perspectives. J Proteomics 2018; 187:93-105. [PMID: 29953962 DOI: 10.1016/j.jprot.2018.06.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/13/2018] [Accepted: 06/23/2018] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is one of the most prevalent malignancies worldwide. Biomarkers that can facilitate better clinical management of CRC are in high demand to improve patient outcome and to reduce mortality. In this regard, proteomic analysis holds a promising prospect in the hunt of novel biomarkers for CRC and in understanding the mechanisms underlying tumorigenesis. This review aims to provide an overview of the current progress of proteomic research, focusing on discovery and validation of diagnostic biomarkers for CRC. We will summarize the contributions of proteomic strategies to recent discoveries of protein biomarkers for CRC and also briefly discuss the potential and challenges of different proteomic approaches in biomarker discovery and translational applications.
Collapse
Affiliation(s)
- Pey Yee Lee
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, 56000 Kuala Lumpur, Malaysia.
| | - Siok-Fong Chin
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, 56000 Kuala Lumpur, Malaysia
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, 56000 Kuala Lumpur, Malaysia
| | - Rahman Jamal
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, 56000 Kuala Lumpur, Malaysia
| |
Collapse
|
22
|
Abdolahi HM, Asiabar AS, Azami-Aghdash S, Pournaghi-Azar F, Rezapour A. Cost-effectiveness of Colorectal Cancer Screening and Treatment Methods: Mapping of Systematic Reviews. Asia Pac J Oncol Nurs 2018; 5:57-67. [PMID: 29379836 PMCID: PMC5763442 DOI: 10.4103/apjon.apjon_50_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/07/2017] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Due to extensive literature on colorectal cancer and their heterogeneous results, this study aimed to summarize the systematic reviews which review the cost-effectiveness studies on different aspects of colorectal cancer. METHODS The required data were collected by searching the following key words according to MeSH: "colorectal cancer," "colorectal oncology," "colorectal carcinoma," "colorectal neoplasm," "colorectal tumors," "cost-effectiveness," "systematic review," and "meta-analysis." The following databases were searched: PubMed, Cochrane, Google Scholar, and Scopus. Two reviewers evaluated the articles according to the checklist of "assessment of multiple systematic reviews" (AMSTAR) tool. RESULTS Finally, eight systematic reviews were included in the study. The Drummond checklist was mostly used for assessing the quality of the articles. The main perspective was related to the payer and the least was relevant to the social. The majority of the cases referred to sensitivity analysis (in 76% of the cases) and the lowest point also was allocated to discounting (in 37% of cases). The Markov model was used most widely in the studies. Treatment methods examined in the studies were not cost-effective in comparison with the studied units. Among the screening methods, computerized tomographic colonography and fecal DNA were cost-effective. The average score of the articles' qualities was high (9.8 out of 11). CONCLUSIONS The community perspective should be taken into consideration at large in the studies. It is necessary to pay more attention to discounting subject in studies. More frequent application of the Markov model is recommended.
Collapse
Affiliation(s)
- Hossein Mashhadi Abdolahi
- Tabriz Health Services Management Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Sarabi Asiabar
- Health Management and Economics Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Saber Azami-Aghdash
- Road Traffic Injury Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Pournaghi-Azar
- Dental and Periodental Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aziz Rezapour
- Health Management and Economics Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Alnabulsi A, Murray GI. Proteomics for early detection of colorectal cancer: recent updates. Expert Rev Proteomics 2017; 15:55-63. [PMID: 29064727 DOI: 10.1080/14789450.2018.1396893] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Colorectal cancer (CRC) is a common type of cancer with a relatively poor survival rate. The survival rate of patients could be improved if CRC is detected early. Biomarkers associated with early stages of tumor development might provide useful tools for the early diagnosis of colorectal cancer. Areas covered: Online searches using PubMed and Google Scholar were performed using keywords and with a focus on recent proteomic studies. The aim of this review is to highlight the need for biomarkers to improve the detection rate of early CRC and provide an overview of proteomic technologies used for biomarker discovery and validation. This review will also discuss recent proteomic studies which focus on identifying biomarkers associated with the early stages of CRC development. Expert commentary: A large number of CRC biomarkers are increasingly being identified by proteomics using diverse approaches. However, the clinical relevance and introduction of these markers into clinical practice cannot be determined without a robust validation process. The size of validation cohorts remains a major limitation in many biomarker studies.
Collapse
Affiliation(s)
- Abdo Alnabulsi
- a Pathology, School of Medicine, Medical Sciences and Nutrition , University of Aberdeen , Aberdeen , UK
| | - Graeme I Murray
- a Pathology, School of Medicine, Medical Sciences and Nutrition , University of Aberdeen , Aberdeen , UK
| |
Collapse
|
24
|
Fan CW, Kuo YB, Lin GP, Chen SM, Chang SH, Li BA, Chan EC. Development of a multiplexed tumor-associated autoantibody-based blood test for the detection of colorectal cancer. Clin Chim Acta 2017; 475:157-163. [PMID: 29074220 DOI: 10.1016/j.cca.2017.10.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/01/2017] [Accepted: 10/22/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common malignancies worldwide, and early diagnosis is vital to improving prognoses. We explored the diagnostic potential of a multiplex autoantibody panel as a biomarker for the detection of CRC by ELISA. METHODS In total, 192 serum samples (92 CRC and 100 matched controls) were tested against a panel of 12 tumor-associated antigens (TAAs): RPH3AL, RPL36, SLP2, p53, survivin, ANAXA4, SEC61B, CCCAP, NYCO16, NMDAR, PLSCR1, and HDAC5. Individual and combined autoantibody signatures were examined. RESULTS Compared to individual autoantibody markers, the combinations of TAAs provided better discrimination between tumorous and normal sera. The overall sensitivity of a selected panel of four antibodies (anti-SLP2, -p53, -SEC61B, and -PLSCR1) was 64.1%, with a specificity of 80% that increased to 83.7% when carcinoembryonic antigen (CEA) measurement was added. Furthermore, the sensitivity of the panel of four antibodies for early and advanced stages of CRC was 66.7% and 62%, increasing to 88.3% and 84%, respectively, when CEA was added. CONCLUSIONS We identified a panel of four antibodies as a promising diagnostic biomarker for the detection of CRC.
Collapse
Affiliation(s)
- Chung-Wei Fan
- Division of Colorectal Surgery, Chang Gung Memorial Hospital, Keelung and Chang Gung University, College of Medicine, Taoyuan, Taiwan
| | | | - Geng-Pin Lin
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Si-Min Chen
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Hsien Chang
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
| | - Bo-An Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Err-Cheng Chan
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan; Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
25
|
Boschetti E, D'Amato A, Candiano G, Righetti PG. Protein biomarkers for early detection of diseases: The decisive contribution of combinatorial peptide ligand libraries. J Proteomics 2017; 188:1-14. [PMID: 28882677 DOI: 10.1016/j.jprot.2017.08.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/09/2017] [Accepted: 08/13/2017] [Indexed: 12/31/2022]
Abstract
The present review deals with biomarker discovery, especially in regard to sample treatment via combinatorial peptide ligand libraries, perhaps the only technique at present allowing deep exploration of biological fluids and tissue extracts in search for low- to very-low-abundance proteins, which could possibly mark the onset of most pathologies. Early-stage biomarkers, in fact, might be the only way to detect the beginning of most diseases thus permitting proper intervention and care. The following cancers are reviewed, with lists of potential biomarkers suggested in various reports: hepatocellular carcinoma, ovarian cancer, breast cancer and pancreatic cancer, together with some other interesting applications. Although panels of proteins have been presented, with robust evidence, as potential early-stage biomarkers in these different pathologies, their approval by FDA as novel biomarkers in routine clinical chemistry settings would require plenty of additional work and efforts from the pharma industry. The science environment in universities could simply not afford such heavy monetary investments. SIGNIFICANCE After more than 16years of search for novel biomarkers, to be used in a clinical chemistry set-up, via proteomic analysis (mostly in biological fluids) it was felt a critical review was due. In the present report, though, only papers reporting biomarker discovery via combinatorial peptide ligand libraries are listed and assessed, since this methodology seems to be the most advanced one for digging in depth into low-to very-low-abundance proteins, which might represent important biomarkers for the onset of pathologies. In particular, a large survey has been made for the following diseases, since they appear to have a large incidence on human population and/or represent fatal diseases: ovarian cancer, breast cancer, pancreatic cancer and hepatocellular carcinoma.
Collapse
Affiliation(s)
| | - Alfonsina D'Amato
- Quadram Institute of Bioscience, Norwich Research Park, NR4 7UA Norwich, UK
| | - Giovanni Candiano
- Nephrology, Dialysis, Transplantation Unit and Laboratory on Pathophysiology of Uremia, Istituto Giannina Gaslini, Genoa, Italy
| | - Pier Giorgio Righetti
- Politecnico di Milano, Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Via Mancinelli 7, Milano 20131, Italy.
| |
Collapse
|
26
|
Singh MP, Rai S, Suyal S, Singh SK, Singh NK, Agarwal A, Srivastava S. Genetic and epigenetic markers in colorectal cancer screening: recent advances. Expert Rev Mol Diagn 2017; 17:665-685. [PMID: 28562109 DOI: 10.1080/14737159.2017.1337511] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Colorectal cancer (CRC) is a heterogenous disease which develops from benign intraepithelial lesions known as adenomas to malignant carcinomas. Acquired alterations in Wnt signaling, TGFβ, MAPK pathway genes and clonal propagation of altered cells are responsible for this transformation. Detection of adenomas or early stage cancer in asymptomatic patients and better prognostic and predictive markers is important for improving the clinical management of CRC. Area covered: In this review, the authors have evaluated the potential of genetic and epigenetic alterations as markers for early detection, prognosis and therapeutic predictive potential in the context of CRC. We have discussed molecular heterogeneity present in CRC and its correlation to prognosis and response to therapy. Expert commentary: Molecular marker based CRC screening methods still fail to gain trust of clinicians. Invasive screening methods, molecular heterogeneity, chemoresistance and low quality test samples are some key challenges which need to be addressed in the present context. New sequencing technologies and integrated omics data analysis of individual or population cohort results in GWAS. MPE studies following a GWAS could be future line of research to establish accurate correlations between CRC and its risk factors. This strategy would identify most reliable biomarkers for CRC screening and management.
Collapse
Affiliation(s)
- Manish Pratap Singh
- a Department of Biotechnology , Motilal Nehru National Institute of Technology (MNNIT) Allahabad , India
| | - Sandhya Rai
- a Department of Biotechnology , Motilal Nehru National Institute of Technology (MNNIT) Allahabad , India
| | - Shradha Suyal
- a Department of Biotechnology , Motilal Nehru National Institute of Technology (MNNIT) Allahabad , India
| | - Sunil Kumar Singh
- a Department of Biotechnology , Motilal Nehru National Institute of Technology (MNNIT) Allahabad , India
| | - Nand Kumar Singh
- a Department of Biotechnology , Motilal Nehru National Institute of Technology (MNNIT) Allahabad , India
| | - Akash Agarwal
- b Department of Surgical Oncology , Dr. Ram Manohar Lohia Institute of Medical Sciences (DRMLIMS) , Lucknow , India
| | - Sameer Srivastava
- a Department of Biotechnology , Motilal Nehru National Institute of Technology (MNNIT) Allahabad , India
| |
Collapse
|
27
|
Garranzo-Asensio M, Guzman-Aranguez A, Povés C, Fernández-Aceñero MJ, Torrente-Rodríguez RM, Ruiz-Valdepeñas Montiel V, Domínguez G, Frutos LS, Rodríguez N, Villalba M, Pingarrón JM, Campuzano S, Barderas R. Toward Liquid Biopsy: Determination of the Humoral Immune Response in Cancer Patients Using HaloTag Fusion Protein-Modified Electrochemical Bioplatforms. Anal Chem 2016; 88:12339-12345. [PMID: 28193070 DOI: 10.1021/acs.analchem.6b03526] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- María Garranzo-Asensio
- Departamento
de Bioquímica y Biología Molecular IV, Facultad de Óptica
y Optometría, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Ana Guzman-Aranguez
- Departamento
de Bioquímica y Biología Molecular IV, Facultad de Óptica
y Optometría, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Carmen Povés
- Gastroenterology
Unit, Hospital Universitario Clínico San Carlos, E-28040 Madrid, Spain
| | | | - Rebeca M. Torrente-Rodríguez
- Departamento
de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Víctor Ruiz-Valdepeñas Montiel
- Departamento
de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Gemma Domínguez
- Departamento de Medicina, Facultad de Medicina, Instituto de Investigaciones Biomédicas “Alberto Sols”, CSIC-UAM, E-28029 Madrid, Spain
| | - Luis San Frutos
- Gynecology
and Obstetrics Department, Hospital Puerta de Hierro, E-28222 Majadahonda, Spain
| | - Nuria Rodríguez
- Medical
Oncology Department, Hospital Universitario La Paz, E-28046 Madrid, Spain
| | - Mayte Villalba
- Departamento
de Bioquímica y Biología Molecular I Facultad de Ciencias
Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - José M. Pingarrón
- Departamento
de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Susana Campuzano
- Departamento
de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Rodrigo Barderas
- Departamento
de Bioquímica y Biología Molecular I Facultad de Ciencias
Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| |
Collapse
|
28
|
Alnabulsi A, Murray GI. Integrative analysis of the colorectal cancer proteome: potential clinical impact. Expert Rev Proteomics 2016; 13:917-927. [PMID: 27598033 DOI: 10.1080/14789450.2016.1233062] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Colorectal cancer (CRC) is one of the common types of cancer that affects a significant proportion of the population and is a major contributor to cancer related mortality. The relatively poor survival rate of CRC could be improved through the identification of clinically useful biomarkers. Areas covered: This review highlights the need for biomarkers and discusses recent proteomics discoveries in the aspects of CRC clinical practice including diagnosis, prognosis, therapy, screening and molecular pathological epidemiology (MPE). Studies have been evaluated in relation to biomarker target, methodology, sample selection, limitations, and potential impact. Finally, the progress in proteomic approaches is briefly discussed and the main difficulties facing the translation of proteomics biomarkers into the clinical practice are highlighted. Expert commentary: The establishment of specific guidelines, best practice recommendations and the improvement in proteomic strategies will significantly improve the prospects for developing clinically useful biomarkers.
Collapse
Affiliation(s)
- Abdo Alnabulsi
- a Pathology, School of Medicine, Medical Sciences and Nutrition , University of Aberdeen , Aberdeen , UK.,b Zoology Building , Vertebrate Antibodies , Aberdeen , UK
| | - Graeme I Murray
- a Pathology, School of Medicine, Medical Sciences and Nutrition , University of Aberdeen , Aberdeen , UK
| |
Collapse
|
29
|
Evaluation of serum nucleoside diphosphate kinase A for the detection of colorectal cancer. Sci Rep 2016; 6:26703. [PMID: 27222072 PMCID: PMC4879623 DOI: 10.1038/srep26703] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 05/09/2016] [Indexed: 01/27/2023] Open
Abstract
We previously described the over-expression of nucleoside diphosphate kinase A (NDKA) in tumours and serum from colorectal cancer (CRC) patients, suggesting its use as biomarker. In this study we evaluated the diagnostic accuracy of serum NDKA to detect advanced neoplasia (CRC or advanced adenomas). Furthermore, the performance of NDKA was compared with the faecal immunochemical test (FIT). The study population included a case-control cohort and a screening cohort (511 asymptomatic first-degree relatives of CRC patients that underwent a colonoscopy and a FIT). Serum NDKA was elevated in CRC patients in the case-control cohort (p = 0.002). In the screening cohort, NDKA levels were higher for advanced adenomas (p = 0.010) and advanced neoplasia (p = 0.006) compared to no neoplasia. Moreover, elevated NDKA was associated with severe characteristics of adenomas (≥3 lesions, size ≥ 1 cm or villous component). Setting specificity to 85%, NDKA showed a sensitivity of 30.19% and 29.82% for advanced adenomas and advanced neoplasia, respectively. NDKA combined with FIT (100 ng/mL cut-off) detected advanced adenomas and advanced neoplasia with 45.28% and 49.12% sensitivity, with specificity close to 90%. The combination of serum NDKA and FIT can improve the detection of advanced neoplasia, mainly for lesions located on the proximal colon, in asymptomatic individuals with CRC family-risk.
Collapse
|