1
|
Almisned FA, Usanase N, Ozsahin DU, Ozsahin I. Incorporation of explainable artificial intelligence in ensemble machine learning-driven pancreatic cancer diagnosis. Sci Rep 2025; 15:14038. [PMID: 40269234 PMCID: PMC12018965 DOI: 10.1038/s41598-025-98298-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
Despite the strides made in medical science, pancreatic cancer continues to be a threat, highlighting the urgent need for creative strategies to address this concern. Recently, a potential approach that has attracted significant attention is using machine learning in clinical decision-making. This research aims to analyze six machine learning algorithms, and an ensemble voting classifier, develop hybrid models for the early detection of pancreatic cancer based on several clinical characteristics and interpret their performance with Shapley Additive Explanations (SHAP). A publicly available dataset composed of 590 patient urine samples was utilized to develop six conventional models for the classification of cancerous from non-cancerous pancreatic cases through the analysis of specific attributes. An ensemble voting classifier was developed from the best-performed single models, which were later hybridized to form six novel hybrid models. The ensemble voting classifier outperformed all stand-alone models with an accuracy of 96.61% and a precision of 98.72%. The six novel hybrid models exhibited higher performance than single models with voting classifier random forest hybridized model outperforming others with an AUC of 99.05% (95% confidence interval (CI): 0.93-1.00) and an interpretation was given by SHAP showing top influential features in pancreatic cancer diagnosis that exhibited the greatest positive SHAP values. Employing rapid sophisticated models with high accuracy and precision holds significant promise in facilitating the effective detection of various diseases, including pancreatic cancer.
Collapse
Affiliation(s)
- Faisal Abdulaziz Almisned
- Department of Information Systems, College of Computer and Information Sciences, King Saud University, Riyadh, Saudi Arabia.
| | - Natacha Usanase
- Operational Research Centre in Healthcare, Near East University, TRNC Mersin 10, Nicosia, 99138, Turkey.
- Department of Biomedical Engineering, Near East University, TRNC Mersin 10, Nicosia, 99138, Turkey.
| | - Dilber Uzun Ozsahin
- Operational Research Centre in Healthcare, Near East University, TRNC Mersin 10, Nicosia, 99138, Turkey
- Department of Medical Diagnostic Imaging, College of Health Science, University of Sharjah, Sharjah, UAE
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, UAE
| | - Ilker Ozsahin
- Operational Research Centre in Healthcare, Near East University, TRNC Mersin 10, Nicosia, 99138, Turkey
| |
Collapse
|
2
|
Kim B, Jung J. Metabolomic Approach to Identify Potential Biomarkers in KRAS-Mutant Pancreatic Cancer Cells. Biomedicines 2024; 12:865. [PMID: 38672219 PMCID: PMC11048406 DOI: 10.3390/biomedicines12040865] [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: 03/28/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Pancreatic cancer is characterized by its high mortality rate and limited treatment options, often driven by oncogenic RAS mutations. In this study, we investigated the metabolomic profiles of pancreatic cancer cells based on their KRAS genetic status. Utilizing both KRAS-wildtype BxPC3 and KRAS-mutant PANC1 cell lines, we identified 195 metabolites differentially altered by KRAS status through untargeted metabolomics. Principal component analysis and hierarchical condition trees revealed distinct separation between KRAS-wildtype and KRAS-mutant cells. Metabolite set enrichment analysis highlighted significant pathways such as homocysteine degradation and taurine and hypotaurine metabolism. Additionally, lipid enrichment analysis identified pathways including fatty acyl glycosides and sphingoid bases. Mapping of identified metabolites to KEGG pathways identified nine significant metabolic pathways associated with KRAS status, indicating diverse metabolic alterations in pancreatic cancer cells. Furthermore, we explored the impact of TRPML1 inhibition on the metabolomic profile of KRAS-mutant pancreatic cancer cells. TRPML1 inhibition using ML-SI1 significantly altered the metabolomic profile, leading to distinct separation between vehicle-treated and ML-SI1-treated PANC1 cells. Metabolite set enrichment analysis revealed enriched pathways such as arginine and proline metabolism, and mapping to KEGG pathways identified 17 significant metabolic pathways associated with TRPML1 inhibition. Interestingly, some metabolites identified in PANC1 compared to BxPC3 were oppositely regulated by TRPML1 inhibition, suggesting their potential as biomarkers for KRAS-mutant cancer cells. Overall, our findings shed light on the distinct metabolite changes induced by both KRAS status and TRPML1 inhibition in pancreatic cancer cells, providing insights into potential therapeutic targets and biomarkers for this deadly disease.
Collapse
Affiliation(s)
| | - Jewon Jung
- Department of SmartBio, College of Life and Health Science, Kyungsung University, Busan 48434, Republic of Korea;
| |
Collapse
|
3
|
Vilà-Quintana L, Fort E, Pardo L, Albiol-Quer MT, Ortiz MR, Capdevila M, Feliu A, Bahí A, Llirós M, Aguilar E, García-Velasco A, Ginestà MM, Laquente B, Pozas D, Lluansí A, Pimenoff VN, Moreno V, Garcia-Gil LJ, Duell EJ, Carreras-Torres R, Aldeguer X. Exploring the Associations of Inflammatory and Oxidative Stress Biomarkers with Pancreatic Diseases: An Observational and Mendelian Randomisation Study. J Clin Med 2024; 13:2247. [PMID: 38673519 PMCID: PMC11050604 DOI: 10.3390/jcm13082247] [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: 03/13/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Identifying biomarkers linked to pancreatic ductal adenocarcinoma (PDAC) and chronic pancreatitis (CP) is crucial for early detection, treatment, and prevention. Methods: Association analyses of 10 serological biomarkers involved in cell signalling (IFN-γ, IL-6, IL-8, IL-10), oxidative stress (superoxide dismutase (SOD) and glutathione peroxidase (GPx) enzyme activities, total glutathione (GSH), malondialdehyde (MDA) levels), and intestinal permeability proteins (zonulin, I-FABP2) were conducted across PDAC (n = 12), CP (n = 21) and control subjects (n = 23). A Mendelian randomisation (MR) approach was used to assess causality of the identified significant associations in two large genetic cohorts (FinnGen and UK Biobank). Results: Observational results showed a downregulation of SOD and GPx antioxidant enzyme activities in PDAC and CP patients, respectively, and higher MDA levels in CP patients. Logistic regression models revealed significant associations between CP and SOD activity (OR = 0.21, 95% CI [0.05, 0.89], per SD), GPx activity (OR = 0.28, 95% CI [0.10, 0.79], per SD), and MDA levels (OR = 2.05, 95% CI [1.36, 3.08], per SD). MR analyses, however, did not support causality. Conclusions: These findings would not support oxidative stress-related biomarkers as potential targets for pancreatic diseases prevention. Yet, further research is encouraged to assess their viability as non-invasive tools for early diagnosis, particularly in pre-diagnostic CP populations.
Collapse
Affiliation(s)
- Laura Vilà-Quintana
- Digestive Diseases and Microbiota Group, Department of Gastroenterology, Girona Biomedical Research Institute (IDIBGI), Hospital Universitari de Girona Dr. Josep Trueta, 17190 Salt, Spain; (L.V.-Q.); (E.F.); (L.P.); (M.C.); (A.F.); (A.B.); (E.A.); (D.P.); (L.J.G.-G.)
| | - Esther Fort
- Digestive Diseases and Microbiota Group, Department of Gastroenterology, Girona Biomedical Research Institute (IDIBGI), Hospital Universitari de Girona Dr. Josep Trueta, 17190 Salt, Spain; (L.V.-Q.); (E.F.); (L.P.); (M.C.); (A.F.); (A.B.); (E.A.); (D.P.); (L.J.G.-G.)
| | - Laura Pardo
- Digestive Diseases and Microbiota Group, Department of Gastroenterology, Girona Biomedical Research Institute (IDIBGI), Hospital Universitari de Girona Dr. Josep Trueta, 17190 Salt, Spain; (L.V.-Q.); (E.F.); (L.P.); (M.C.); (A.F.); (A.B.); (E.A.); (D.P.); (L.J.G.-G.)
| | - Maria T. Albiol-Quer
- General and Digestive Surgery Group, Department of Surgery, Girona Biomedical Research Institute (IDIBGI), Hospital Universitari de Girona Dr. Josep Trueta, 17007 Girona, Spain;
| | - Maria Rosa Ortiz
- Department of Pathology, Hospital Universitari de Girona Dr. Josep Trueta, 17007 Girona, Spain;
| | - Montserrat Capdevila
- Digestive Diseases and Microbiota Group, Department of Gastroenterology, Girona Biomedical Research Institute (IDIBGI), Hospital Universitari de Girona Dr. Josep Trueta, 17190 Salt, Spain; (L.V.-Q.); (E.F.); (L.P.); (M.C.); (A.F.); (A.B.); (E.A.); (D.P.); (L.J.G.-G.)
| | - Anna Feliu
- Digestive Diseases and Microbiota Group, Department of Gastroenterology, Girona Biomedical Research Institute (IDIBGI), Hospital Universitari de Girona Dr. Josep Trueta, 17190 Salt, Spain; (L.V.-Q.); (E.F.); (L.P.); (M.C.); (A.F.); (A.B.); (E.A.); (D.P.); (L.J.G.-G.)
| | - Anna Bahí
- Digestive Diseases and Microbiota Group, Department of Gastroenterology, Girona Biomedical Research Institute (IDIBGI), Hospital Universitari de Girona Dr. Josep Trueta, 17190 Salt, Spain; (L.V.-Q.); (E.F.); (L.P.); (M.C.); (A.F.); (A.B.); (E.A.); (D.P.); (L.J.G.-G.)
| | - Marc Llirós
- Bioinformatics and Bioimaging (BI-SQUARED) Research Group, Biosciences Department, Faculty of Sciences, Technology and Engineerings, Universitat de Vic—Universitat Central de Catalunya, 08500 Vic, Spain;
| | - Esther Aguilar
- Digestive Diseases and Microbiota Group, Department of Gastroenterology, Girona Biomedical Research Institute (IDIBGI), Hospital Universitari de Girona Dr. Josep Trueta, 17190 Salt, Spain; (L.V.-Q.); (E.F.); (L.P.); (M.C.); (A.F.); (A.B.); (E.A.); (D.P.); (L.J.G.-G.)
| | - Adelaida García-Velasco
- Precision Oncology Group, Girona Biomedical Research Institute (IDIBGI), Institut Català d’Oncologia (ICO), Hospital Universitari de Girona Dr. Josep Trueta, 17007 Girona, Spain;
| | - Mireia M. Ginestà
- Hereditary Cancer Program, Oncobell Program, CIBERONC, Institut Català d’Oncologia (ICO), Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), 08908 Barcelona, Spain;
| | - Berta Laquente
- Medical Oncology Department, Institut Català d’Oncologia (ICO), Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), 08908 Barcelona, Spain;
| | - Débora Pozas
- Digestive Diseases and Microbiota Group, Department of Gastroenterology, Girona Biomedical Research Institute (IDIBGI), Hospital Universitari de Girona Dr. Josep Trueta, 17190 Salt, Spain; (L.V.-Q.); (E.F.); (L.P.); (M.C.); (A.F.); (A.B.); (E.A.); (D.P.); (L.J.G.-G.)
| | - Aleix Lluansí
- Institut de Recerca Sant Joan de Déu (IRSJD), Hospital Sant Joan de Déu, 08950 Barcelona, Spain;
| | - Ville Nikolai Pimenoff
- Department of Clinical Science, Intervention and Technology—CLINTEC, Karolinska Institutet, 141 52 Huddinge, Sweden;
- Unit of Population Health, Faculty of Medicine, University of Oulu, 90570 Oulu, Finland
| | - Victor Moreno
- Catalan Institute of Oncology (ICO), Institut de Recerca Biomedica de Bellvitge (IDIBELL), 08908 Barcelona, Spain;
- Department of Clinical Sciences, Faculty of Medicine and health Sciences and Universitat de Barcelona Institute of Complex Systems (UBICS), University of Barcelona (UB), L’Hospitalet de Llobregat, 08028 Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
| | - Libadro Jesús Garcia-Gil
- Digestive Diseases and Microbiota Group, Department of Gastroenterology, Girona Biomedical Research Institute (IDIBGI), Hospital Universitari de Girona Dr. Josep Trueta, 17190 Salt, Spain; (L.V.-Q.); (E.F.); (L.P.); (M.C.); (A.F.); (A.B.); (E.A.); (D.P.); (L.J.G.-G.)
| | - Eric J. Duell
- Cancer Epidemiology Research Program, Unit of Nutrition and Cancer, Institut Català d’Oncologia (ICO), Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), 08908 Barcelona, Spain;
| | - Robert Carreras-Torres
- Digestive Diseases and Microbiota Group, Department of Gastroenterology, Girona Biomedical Research Institute (IDIBGI), Hospital Universitari de Girona Dr. Josep Trueta, 17190 Salt, Spain; (L.V.-Q.); (E.F.); (L.P.); (M.C.); (A.F.); (A.B.); (E.A.); (D.P.); (L.J.G.-G.)
| | - Xavier Aldeguer
- Digestive Diseases and Microbiota Group, Department of Gastroenterology, Girona Biomedical Research Institute (IDIBGI), Hospital Universitari de Girona Dr. Josep Trueta, 17190 Salt, Spain; (L.V.-Q.); (E.F.); (L.P.); (M.C.); (A.F.); (A.B.); (E.A.); (D.P.); (L.J.G.-G.)
| |
Collapse
|
4
|
Attebury H, Daley D. The Gut Microbiome and Pancreatic Cancer Development and Treatment. Cancer J 2023; 29:49-56. [PMID: 36957973 PMCID: PMC10042586 DOI: 10.1097/ppo.0000000000000647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
ABSTRACT Changes in the gut microbiome have been increasingly shown to accompany oncogenesis across various tumors. Similarly, microbial dysbiosis was found to be associated with pancreatic cancer progression and survival outcomes, expanding the field of tumor microenvironment research in pancreatic cancer. Mechanistic studies in pancreatic cancer models implicate components of the gut and pancreatic cancer microbiome in regulating tumorigenesis by altering cancer cell signaling, modulating immune function, and influencing the efficacy of current therapies in pancreatic cancer. This review discusses the outcomes of microbial modulation across various preclinical and clinical studies and highlights ongoing trials targeting the microbiome for pancreatic cancer therapy.
Collapse
|
5
|
Characterization of Mesothelin Glycosylation in Pancreatic Cancer: Decreased Core Fucosylated Glycoforms in Pancreatic Cancer Patients’ Sera. Biomedicines 2022; 10:biomedicines10081942. [PMID: 36009489 PMCID: PMC9405996 DOI: 10.3390/biomedicines10081942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/27/2022] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
Abstract
Currently, there are no reliable biomarkers for the diagnosis of pancreatic cancer (PaC). Glycoproteomic approaches that analyze the glycan determinants on specific glycoproteins have proven useful to develop more specific cancer biomarkers than the corresponding protein levels. In PaC, mesothelin (MSLN) is a neo-expressed glycoprotein. MSLN glycosylation has not been described and could be altered in PaC. In this work, we aimed to characterize MSLN glycans from PaC cells and serum samples to assess their potential usefulness as PaC biomarkers. First, we analyzed MSLN glycans from PaC cell lines and then we developed an enzyme-linked lectin assay to measure core fucosylated-MSLN (Cf-MSLN) glycoforms. MSLN glycans from PaC cells were analyzed by glycan sequencing and through Western blotting with lectins. All of the cell lines secreted MSLN, with its three N-glycosylation sites occupied by complex-type N-glycans, which were mainly α2,3-sialylated, core fucosylated and highly branched. The Cf-MSLN glycoforms were quantified on PaC serum samples, and compared with MSLN protein levels. The Cf-MSLN was significantly decreased in PaC patients compared to control sera, while no differences were detected by using MSLN protein levels. In conclusion, Cf-MSLN glycoforms were differently expressed in PaC, which opens the way to further investigate their usefulness as PaC biomarkers.
Collapse
|
6
|
Phaseolus vulgaris Erythroagglutinin (PHA-E)-Positive Ceruloplasmin Acts as a Potential Biomarker in Pancreatic Cancer Diagnosis. Cells 2022; 11:cells11152453. [PMID: 35954297 PMCID: PMC9367852 DOI: 10.3390/cells11152453] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/31/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022] Open
Abstract
Pancreatic cancer (PC) remains one of the top 10 causes of cancer-related death in recent years. Approximately 80% of PC patients are diagnosed at the middle or advanced stage and miss the opportunity for surgery. The demand for early diagnostic methods and reliable biomarkers is increasing, although a number of tumor markers such as CA19-9 and CEA have already been utilized in clinics. In this study, we analyzed the alteration of N-glycan of serum glycoproteins by mass spectrometry and lectin blotting. The results showed that bisecting GlcNAc structures of glycoproteins are significantly increased in PC patients' sera. With Phaseolus vulgaris Erythroagglutinin (PHA-E) lectin that specifically recognizes bisecting GlcNAc N-glycans, the serum glycoproteins bearing bisecting GlcNAc in PC patients' sera were pulled down and identified by nano-LC-MS/MS. Among them, ceruloplasmin (Cp) was screened out with a satisfied sensitivity and specificity in identifying PC from acute pancreatitis patients (AUC: 0.757) and normal healthy persons (AUC: 0.972), suggesting a close association between Cp and PC development and diagnosis. To prove that, the Cp expression in tumor tissues of PC patients was examined. The results showed that Cp was significantly upregulated in PC tissues compared to that in adjacent normal tissues. All these results suggested that PHA-E-positive Cp could be a potential PC-specific glycoprotein marker to distinguish PC patients from acute pancreatitis patients and normal persons.
Collapse
|
7
|
Boyer S, Lee HJ, Steele N, Zhang L, Sajjakulnukit P, Andren A, Ward MH, Singh R, Basrur V, Zhang Y, Nesvizhskii AI, Pasca di Magliano M, Halbrook CJ, Lyssiotis CA. Multiomic characterization of pancreatic cancer-associated macrophage polarization reveals deregulated metabolic programs driven by the GM-CSF-PI3K pathway. eLife 2022; 11:e73796. [PMID: 35156921 PMCID: PMC8843093 DOI: 10.7554/elife.73796] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
The pancreatic ductal adenocarcinoma microenvironment is composed of a variety of cell types and marked by extensive fibrosis and inflammation. Tumor-associated macrophages (TAMs) are abundant, and they are important mediators of disease progression and invasion. TAMs are polarized in situ to a tumor promoting and immunosuppressive phenotype via cytokine signaling and metabolic crosstalk from malignant epithelial cells and other components of the tumor microenvironment. However, the specific distinguishing features and functions of TAMs remain poorly defined. Here, we generated tumor-educated macrophages (TEMs) in vitro and performed detailed, multiomic characterization (i.e., transcriptomics, proteomics, metabolomics). Our results reveal unique genetic and metabolic signatures of TEMs, the veracity of which were queried against our in-house single-cell RNA sequencing dataset of human pancreatic tumors. This analysis identified expression of novel, metabolic TEM markers in human pancreatic TAMs, including ARG1, ACLY, and TXNIP. We then utilized our TEM model system to study the role of mutant Kras signaling in cancer cells on TEM polarization. This revealed an important role for granulocyte-macrophage colony-stimulating factor (GM-CSF) and lactate on TEM polarization, molecules released from cancer cells in a mutant Kras-dependent manner. Lastly, we demonstrate that GM-CSF dysregulates TEM gene expression and metabolism through PI3K-AKT pathway signaling. Collectively, our results define new markers and programs to classify pancreatic TAMs, how these are engaged by cancer cells, and the precise signaling pathways mediating polarization.
Collapse
Affiliation(s)
- Seth Boyer
- Department of Molecular & Integrative Physiology, University of MichiganAnn ArborUnited States
| | - Ho-Joon Lee
- Department of Molecular & Integrative Physiology, University of MichiganAnn ArborUnited States
| | - Nina Steele
- Department of Cell and Developmental Biology, University of MichiganAnn ArborUnited States
- Department of Surgery, University of MichiganAnn ArborUnited States
| | - Li Zhang
- Department of Molecular & Integrative Physiology, University of MichiganAnn ArborUnited States
| | - Peter Sajjakulnukit
- Department of Molecular & Integrative Physiology, University of MichiganAnn ArborUnited States
| | - Anthony Andren
- Department of Molecular & Integrative Physiology, University of MichiganAnn ArborUnited States
| | - Matthew H Ward
- Department of Molecular & Integrative Physiology, University of MichiganAnn ArborUnited States
| | - Rima Singh
- Department of Molecular Biology and Biochemistry, University of California, IrvineIrvineUnited States
| | - Venkatesha Basrur
- Department of Pathology, University of MichiganAnn ArborUnited States
| | - Yaqing Zhang
- Department of Surgery, University of MichiganAnn ArborUnited States
| | - Alexey I Nesvizhskii
- Department of Pathology, University of MichiganAnn ArborUnited States
- Department of Computational Medicine and Bioinformatics, University of MichiganAnn ArborUnited States
| | - Marina Pasca di Magliano
- Department of Surgery, University of MichiganAnn ArborUnited States
- Rogel Cancer Center, University of MichiganAnn ArborUnited States
| | - Christopher J Halbrook
- Department of Molecular & Integrative Physiology, University of MichiganAnn ArborUnited States
| | - Costas A Lyssiotis
- Department of Molecular & Integrative Physiology, University of MichiganAnn ArborUnited States
- Rogel Cancer Center, University of MichiganAnn ArborUnited States
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of MichiganAnn ArborUnited States
| |
Collapse
|
8
|
Zhang J, Wei Q, Dong D, Ren L. The role of TPS, CA125, CA15-3 and CEA in prediction of distant metastasis of breast cancer. Clin Chim Acta 2021; 523:19-25. [PMID: 34454906 DOI: 10.1016/j.cca.2021.08.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To explore the application value of breast cancer tumor markers tissue polypeptide specific antigen (TPS), carbohydrate antigen 15-3 (CA15-3), carcinoembryonic antigen (CEA), carbohydrate antigen 125 (CA125) detection alone or in combination for the monitoring of distant metastasis of breast cancer. METHOD The clinical data of 389 female breast cancer patients admitted to Tianjin Medical University Cancer Institute and Hospital from January 2016 to March 2017 were retrospectively analyzed. Serum levels of TPS, CA125, CA15-3, and CEA were compared to analyze their significance in prediction distant metastasis of breast cancer. The patients were divided into the distant metastatic group and the non-metastatic group according to whether the patients had distant metastasis. The non-metastatic group was divided into the control group and the occult metastasis group according to whether distant metastases occurred in 3 years after treatment. RESULT The receiver operating characteristic curve analysis revealed that all four markers had the diagnostic value in distant metastasis of breast cancer (AUCTPS = 0.754, AUC15-3 = 0.821, AUCCEA = 0.755, AUCCA125 = 0.651) and in occult metastasis in 3 years after treatment (AUCTPS = 0.751, AUC15-3 = 0.744, AUCCEA = 0.725, AUCCA125 = 0.661). To estimate whether the discrimination ability could be improved by marker panels, we established marker panels composed of TPS, CA125, CA15-3, and CEA. To discriminate distant metastasis from non-distant metastasis, the diagnostic ability of different panels composed of TPS, CA125, CA15-3 and CEA did not show significant difference compared with single CA15-3 (P > 0.05). To discriminate occult metastasis from the control group, no significant difference was shown in AUC between marker panels and single marker (P > 0.05). However, the sensitivity was improved when the marker-panels were used overall. CONCLUSION All tumor markers have abilities in prediction of distant metastasis of breast cancer. The combined detection of the markers is more valuable than using single tumor marker in improving sensitivity. Two markers' panels are more suitable for the prediction of distant metastasis of breast cancer than three or four makers' panels with the similar sensitivity and AUC.
Collapse
Affiliation(s)
- Jingya Zhang
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, PR China
| | - Qian Wei
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, PR China
| | - Dong Dong
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, PR China
| | - Li Ren
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, PR China.
| |
Collapse
|
9
|
Non-Invasive Biomarkers for Earlier Detection of Pancreatic Cancer-A Comprehensive Review. Cancers (Basel) 2021; 13:cancers13112722. [PMID: 34072842 PMCID: PMC8198035 DOI: 10.3390/cancers13112722] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC), which represents approximately 90% of all pancreatic cancers, is an extremely aggressive and lethal disease. It is considered a silent killer due to a largely asymptomatic course and late clinical presentation. Earlier detection of the disease would likely have a great impact on changing the currently poor survival figures for this malignancy. In this comprehensive review, we assessed over 4000 reports on non-invasive PDAC biomarkers in the last decade. Applying the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool, we selected and reviewed in more detail 49 relevant studies reporting on the most promising candidate biomarkers. In addition, we also highlight the present challenges and complexities of translating novel biomarkers into clinical use. Abstract Pancreatic ductal adenocarcinoma (PDAC) carries a deadly diagnosis, due in large part to delayed presentation when the disease is already at an advanced stage. CA19-9 is currently the most commonly utilized biomarker for PDAC; however, it lacks the necessary accuracy to detect precursor lesions or stage I PDAC. Novel biomarkers that could detect this malignancy with improved sensitivity (SN) and specificity (SP) would likely result in more curative resections and more effective therapeutic interventions, changing thus the present dismal survival figures. The aim of this study was to systematically and comprehensively review the scientific literature on non-invasive biomarkers in biofluids such as blood, urine and saliva that were attempting earlier PDAC detection. The search performed covered a period of 10 years (January 2010—August 2020). Data were extracted using keywords search in the three databases: MEDLINE, Web of Science and Embase. The Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool was applied for study selection based on establishing the risk of bias and applicability concerns in Patient Selection, Index test (biomarker assay) and Reference Standard (standard-of-care diagnostic test). Out of initially over 4000 published reports, 49 relevant studies were selected and reviewed in more detail. In addition, we discuss the present challenges and complexities in the path of translating the discovered biomarkers into the clinical setting. Our systematic review highlighted several promising biomarkers that could, either alone or in combination with CA19-9, potentially improve earlier detection of PDAC. Overall, reviewed biomarker studies should aim to improve methodological and reporting quality, and novel candidate biomarkers should be investigated further in order to demonstrate their clinical usefulness. However, challenges and complexities in the path of translating the discovered biomarkers from the research laboratory to the clinical setting remain and would have to be addressed before a more realistic breakthrough in earlier detection of PDAC is achieved.
Collapse
|
10
|
Zhong C, Li ZX, Yang LJ, Wu G, Xiang B, Wang YL, Zhou Q. ITGB6 may promote proliferation and invasion in pancreatic cancer. Arch Med Sci 2021; 20:267-279. [PMID: 38414469 PMCID: PMC10895961 DOI: 10.5114/aoms/114039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 11/06/2019] [Indexed: 02/29/2024] Open
Abstract
Introduction The ITGB6 gene encoding a protein that can regulate the integrin αvβ6 heterodimer protein expression in different status was shown to play an important role in multiple human cancers, such as brain cancer, colon cancer and oral cancer, and is related to clinical progression. This study aims to explore the function and the mechanism of the ITGB6 gene or protein in pancreatic cancer. Material and methods We examined the expression of ITGB6 in pancreatic cancer using immunohistochemistry and analyzed the relationship between the expression of ITGB6 and the clinicopathologic features in pancreatic cancer patients. In addition, a bioinformatic method was used to analyze the ITGB6 mRNA level in pancreatic tumor tissues compared with normal pancreatic tissues and to analyze the correlation between high KIF23 expression and prognosis in pancreatic cancer patients. Moreover, colony formation assay, MTT assay, cell scratch, cell invasion and western blot assays in vitro and a xenograft mouse model in vivo were performed to analyze the effect of KIF23 on proliferation and invasion of pancreatic cancer cells. Results Increased expression of ITGB6 was significantly correlated with poor clinical outcome in both our clinical data and TCGA data of pancreatic cancer. Furthermore, functional assays revealed that ITGB6 knockdown in vivo and in vitro might inhibit cancer cell proliferation and the ability of invasion or migration. Conclusions Our data suggest that ITGB6 is associated with pancreatic cancer malignant progression. Hence, ITGB6 may serve as a potential target of pancreatic cancer for future research, and further study is needed.
Collapse
Affiliation(s)
- Chao Zhong
- Department of Traditional Chinese Medicine of Orthopedic and Traumatic, Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu City, Sichuan Province, China
| | - Zhi-Xi Li
- Department of Respiratory Medicine, East Hospital, Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu City, Sichuan Province, China
| | - Ling-Jing Yang
- Department of Respiratory Medicine, East Hospital, Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu City, Sichuan Province, China
| | - Gang Wu
- Department of Hepatobiliary Surgery, Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu City, Sichuan Province, China
| | - Bo Xiang
- Department of Cardiosurgery, Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu City, Sichuan Province, China
| | - Yu-Lan Wang
- Department of Oncology, Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu City, Sichuan Province, China
| | - Qing Zhou
- Department of Ultrasound, Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu City, Sichuan Province, China
| |
Collapse
|
11
|
Varvanina G, Lesko K, Bordin D, Dubtsova E, Malykh M, Noskova K, Vinokurova L. Blood biomarkers and computed tomography for differential diagnosis of pancreatic cancer and chronic pancreatitis. DOKAZATEL'NAYA GASTROENTEROLOGIYA 2021; 10:12. [DOI: 10.17116/dokgastro20211004112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
|
12
|
Liu W, Tang J, Zhang H, Kong F, Zhu H, Li P, Li Z, Kong X, Wang K. A novel lncRNA PTTG3P/miR-132/212-3p/FoxM1 feedback loop facilitates tumorigenesis and metastasis of pancreatic cancer. Cell Death Discov 2020; 6:136. [PMID: 33298873 PMCID: PMC7705684 DOI: 10.1038/s41420-020-00360-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 12/19/2022] Open
Abstract
Pseudogene pituitary tumor-transforming 3 (PTTG3P) is emerging as a key player in the development and progression of cancer. However, the biological role and clinical significance of PTTG3P in pancreatic ductal adenocarcinoma (PDAC) remain unclear. Here, we found that PTTG3P was significantly upregulated in PDAC tissues. Elevated PTTG3P expression correlated with larger tumor size and worse differentiation, and reduced overall survival. Bioinformatics and experimental evidence revealed that PTTG3P promoted malignant phenotypes and FoxM1 signaling pathway in PDAC cells. Mechanistically, PTTG3P functions as a microRNA sponge to positively regulate the expression of FoxM1 through sponging miR-132/212-3p. Moreover, it showed that FoxM1 transcriptionally activated PTTG3P expression, thus forming a feedback loop to promote the aggressiveness of PDAC cells. Taken together, our findings suggest that PTTG3P promotes PDAC progression through PTTG3P/miR-132/212-3p/FoxM1 feedforward circuitry and it may serve as a promising diagnostic marker or target for treatment in PDAC patients.
Collapse
Affiliation(s)
- Wenyu Liu
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.,Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Jian Tang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Huiqing Zhang
- Department of Gastrointestinal Medical Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, China
| | - Fanyang Kong
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Huiyun Zhu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Ping Li
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Zhaoshen Li
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| | - Xiangyu Kong
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| | - Kaixuan Wang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
13
|
Guerrero PE, Duran A, Ortiz MR, Castro E, Garcia-Velasco A, Llop E, Peracaula R. Microfibril associated protein 4 (MFAP4) is a carrier of the tumor associated carbohydrate sialyl-Lewis x (sLe x) in pancreatic adenocarcinoma. J Proteomics 2020; 231:104004. [PMID: 33038510 DOI: 10.1016/j.jprot.2020.104004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/04/2020] [Accepted: 10/04/2020] [Indexed: 02/07/2023]
Abstract
Late diagnosis of pancreatic ductal adenocarcinoma (PDA) is one of the reasons of its low 5-year survival rate and it is due to its unspecific symptoms during the first stages of the disease and the lack of reliable serological markers. Since PDA shows an altered glycan expression, here we have focused on finding novel potential biomarkers, namely glycoproteins that express the tumor associated carbohydrate structure sialyl-Lewis x (sLex), which is described in PDA. Through a glycoproteomic approach, we have analyzed target proteins containing sLex from PDA tissues by 2DE and immunodetection techniques, and have identified by mass spectrometry the protein MFAP4 as a carrier of sLex in PDA. MFAP4 showed a higher expression in PDA tissues compared with pancreatic control tissues. In addition, the colocalization of sLex over MFAP4 was found only in PDA and not in control pancreatic tissues. The analysis of MFAP4 expression in PDA cell lines and their secretome, in combination with immunohistochemistry of pancreatic tissues, revealed that MFAP4 was not produced by PDA cells, but it was found in the pancreatic extracellular matrix. The specificity of MFAP4 glycoform containing sLex in PDA tissues shows its relevance as a potential PDA biomarker. SIGNIFICANCE: Despite advances in the field of cancer research, pancreatic ductal adenocarcinoma (PDA) lacks of a specific and sensitive biomarker for its early detection, when curative resection is still possible before metastases arise. Thus, efforts to discover new PDA biomarkers represent the first line in the fight against the increase of its incidence reported in recent years. Glycan alterations on glycoconjugates, such as glycoproteins have emerged as a rich source for the identification of novel cancer markers. In the present work, we aimed to shed light on novel biomarkers based on altered glycosylation in PDA, in particular those glycoproteins of PDA tissues carrying the tumor carbohydrate antigen sialyl-Lewis x (sLex). Through a glycoproteomic approach, we have shown that the glycoprotein MFAP4 carries sLex in PDA tissues and not in control pancreatic tissues. MFAP4 is found in the extracellular matrix in PDA and although its role in cancer progression is unclear, its sLex glycoform could be a potential biomarker in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Pedro Enrique Guerrero
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, 17003 Girona, Spain
| | - Adrià Duran
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, 17003 Girona, Spain
| | - Maria Rosa Ortiz
- Pathology Department, Josep Trueta University Hospital, 17007 Girona, Spain
| | - Ernesto Castro
- Hepato-biliary and Pancreatic Surgery Unit, Josep Trueta University Hospital, 17007 Girona, Spain
| | | | - Esther Llop
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, 17003 Girona, Spain..
| | - Rosa Peracaula
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, 17003 Girona, Spain..
| |
Collapse
|
14
|
Singh RR, Mohammad J, Orr M, Reindl KM. Glutathione S-Transferase pi-1 Knockdown Reduces Pancreatic Ductal Adenocarcinoma Growth by Activating Oxidative Stress Response Pathways. Cancers (Basel) 2020; 12:E1501. [PMID: 32526885 PMCID: PMC7352757 DOI: 10.3390/cancers12061501] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022] Open
Abstract
Glutathione S-transferase pi-1 (GSTP1) plays an important role in regulating oxidative stress by conjugating glutathione to electrophiles. GSTP1 is overexpressed in breast, colon, lung, and prostate tumors, where it contributes to tumor progression and drug resistance; however, the role of GSTP1 in pancreatic ductal adenocarcinoma (PDAC) is not well understood. Using shRNA, we knocked down GSTP1 expression in three different PDAC cell lines and determined the effect on cell proliferation, cell cycle progression, and reactive oxygen species (ROS) levels. Our results show GSTP1 knockdown reduces PDAC cell growth, prolongs the G0/G1 phase, and elevates ROS in PDAC cells. Furthermore, GSTP1 knockdown results in the increased phosphorylation of c-Jun N-terminal kinase (JNK) and c-Jun and the decreased phosphorylation of extracellular signal-regulated kinase (ERK), p65, the reduced expression of specificity protein 1 (Sp1), and the increased expression of apoptosis-promoting genes. The addition of the antioxidant glutathione restored cell viability and returned protein expression levels to those found in control cells. Collectively, these data support the working hypothesis that the loss of GSTP1 elevates oxidative stress, which alters mitogen-activated protein (MAP) kinases and NF-κB signaling, and induces apoptosis. In support of these in vitro data, nude mice bearing orthotopically implanted GSTP1-knockdown PDAC cells showed an impressive reduction in the size and weight of tumors compared to the controls. Additionally, we observed reduced levels of Ki-67 and increased expression of cleaved caspase-3 in GSTP1-knockdown tumors, suggesting GSTP1 knockdown impedes proliferation and upregulates apoptosis in PDAC cells. Together, these results indicate that GSTP1 plays a significant role in PDAC cell growth and provides support for the pursuit of GSTP1 inhibitors as therapeutic agents for PDAC.
Collapse
Affiliation(s)
- Rahul R. Singh
- Department of Biological Sciences, North Dakota State University, Fargo, ND 58108, USA; (R.R.S.); (J.M.)
| | - Jiyan Mohammad
- Department of Biological Sciences, North Dakota State University, Fargo, ND 58108, USA; (R.R.S.); (J.M.)
| | - Megan Orr
- Department of Statistics, North Dakota State University, Fargo, ND 58108, USA;
| | - Katie M. Reindl
- Department of Biological Sciences, North Dakota State University, Fargo, ND 58108, USA; (R.R.S.); (J.M.)
| |
Collapse
|
15
|
An Immunosensor for the Detection of ULBP2 Biomarker. MICROMACHINES 2020; 11:mi11060568. [PMID: 32503144 PMCID: PMC7344431 DOI: 10.3390/mi11060568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 01/20/2023]
Abstract
Pancreatic cancer (PC) is a global health problem that features a very high mortality rate. The UL16 binding protein 2 (ULBP2) is a new biomarker for PC detection. This study develops a simple, reliable, and inexpensive immunosensor for the detection of the ULBP2 antigen while also investigating the effects of an array configuration of connected sensors and zinc oxide (ZnO) nanoparticles on the immunosensor’s sensitivity. The ULBP2 antibody was immobilized onto the screen-printed carbon electrode (SPCE) surfaces of three different sensors: a simple SPCE (ULBP2-SPCE); an SPCE array, which is a series of identical SPCE connected to each other at different arrangements of rows and columns (ULBP2-SPCE-1x2 and ULBP2-SPCE-1x3); and an SPCE combined with ZnO nanoparticles (ULBP2-ZnO/SPCE). Impedance spectrum measurements for the immunosensors to ULBP2 antigen were conducted and compared. According to the result, the array configurations (ULBP2-SPCE-1x2 and ULBP2-SPCE-1x3) show an improvement of sensitivity compared to the ULBP2-SPCE alone, but the improvement is not as significant as that of the ULBP2-ZnO/SPCE configuration (ULBP2-ZnO/SPCE > ULBP2-SPCE: 18 times larger). The ULBP2-ZnO/SPCE immunosensor has a low limit of detection (1 pg/mL) and a high sensitivity (332.2 Ω/Log(pg/mL)), excellent linearity (R2 = 0.98), good repeatability (coefficients of variation = 5.03%), and is stable in long-term storage (retaining 95% activity after 28 days storage). In an array configuration, the immunosensor has an increased signal-to-noise ratio (ULBP2-SPCE-1x3 > ULBP2-SPCE: 1.5-fold) and sensitivity (ULBP2-SPCE-1x3 > ULBP2-SPCE: 2.6-fold). In conclusion, either the modification with ZnO nanoparticles onto the sensor or the use of an array configuration of sensors can enhance the immunosensor’s sensitivity. In this study, the best immunosensor for detecting ULBP2 antigens is the ULBP2-ZnO/SPCE immunosensor.
Collapse
|
16
|
Arpalahti L, Haglund C, Holmberg CI. Proteostasis Dysregulation in Pancreatic Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1233:101-115. [PMID: 32274754 DOI: 10.1007/978-3-030-38266-7_4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
The most common form of pancreatic cancer, pancreatic ductal adenocarcinoma (PDAC), has a dismal 5-year survival rate of less than 5%. Radical surgical resection, in combination with adjuvant chemotherapy, provides the best option for long-term patient survival. However, only approximately 20% of patients are resectable at the time of diagnosis, due to locally advanced or metastatic disease. There is an urgent need for the identification of new, specific, and more sensitive biomarkers for diagnosis, prognosis, and prediction to improve the treatment options for pancreatic cancer patients. Dysregulation of proteostasis is linked to many pathophysiological conditions, including various types of cancer. In this review, we report on findings relating to the main cellular protein degradation systems, the ubiquitin-proteasome system (UPS) and autophagy, in pancreatic cancer. The expression of several components of the proteolytic network, including E3 ubiquitin-ligases and deubiquitinating enzymes, are dysregulated in PDAC, which accounts for approximately 90% of all pancreatic malignancies. In the future, a deeper understanding of the emerging role of proteostasis in pancreatic cancer has the potential to provide clinically relevant biomarkers and new strategies for combinatorial therapeutic options to better help treat the patients.
Collapse
Affiliation(s)
- Leena Arpalahti
- Medicum, Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland
| | - Caj Haglund
- Research Programs Unit, Translational Cancer Medicine Program, University of Helsinki, Helsinki, Finland
- Department of Surgery, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Carina I Holmberg
- Medicum, Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
17
|
Liu ZH, Dong SX, Jia JH, Zhang ZL, Zhen ZG. KIF3B Promotes the Proliferation of Pancreatic Cancer. Cancer Biother Radiopharm 2019; 34:355-361. [PMID: 31157987 DOI: 10.1089/cbr.2018.2716] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Zhi-Hu Liu
- Hepatobiliary Surgery, Xingtai People's Hospital, The Affiliated Hospital of Hebei Medical University, Xingtai City, China
| | - Shu-Xiao Dong
- Department of Obstetrics, The Third People's Hospital in Xingtai City, Xingtai City, China
| | - Jun-Hong Jia
- Hepatobiliary Surgery, Xingtai People's Hospital, The Affiliated Hospital of Hebei Medical University, Xingtai City, China
| | - Zhen-Liang Zhang
- Hepatobiliary Surgery, Xingtai People's Hospital, The Affiliated Hospital of Hebei Medical University, Xingtai City, China
| | - Zhong-Guang Zhen
- Hepatobiliary Surgery, Xingtai People's Hospital, The Affiliated Hospital of Hebei Medical University, Xingtai City, China
| |
Collapse
|
18
|
Magnetized Carbon Nanotube Based Lateral Flow Immunoassay for Visual Detection of Complement Factor B. Molecules 2019; 24:molecules24152759. [PMID: 31366012 PMCID: PMC6695926 DOI: 10.3390/molecules24152759] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/23/2019] [Accepted: 07/26/2019] [Indexed: 01/02/2023] Open
Abstract
The authors describe a magnetized carbon nanotube (MCNT) based lateral flow immunoassay (LFI) for visual detection of complement factor B (CFB) in blood. MCNT was prepared by decorating magnetic Fe3O4 nanoparticles on multi-walled CNT surface and used as a colored tag for LFI. Monoclonal antibody (mAb, Ab1) of CFB was covalently immobilized on the MCNT surface via diimide-activated conjugation between the carboxyl groups on the MCNT surface and amino groups of antibodies. Polyclonal antibody of CFB (Ab2) and the secondary antibody were used to prepare the lateral flow test strips. The assay involved: (1) the capture of CFB in blood with the mAb-functionalized MCNT; (2) magnetic separation of the formed CFB-mAb-MCNT and excess of mAb-MCNT from the blood with an external magnet; (3) lateral flow test to capture the CFB-mAb-MCNT complex on the test zone and the excess of mAb-MCNT on the control zone; (4) Recording the intensities of the produced the characteristic brown bands with a portable strip reader and quantitating the concentration of CFB. The proof-of-concept was demonstrated by testing CFB in the buffer, and the detection limit was 5 ng mL-1 under the optimized analytical parameters. CFB in 1 μL of human blood was detected successfully in 30 min with this LFI and the results had a high correlation with commercial ELISA kit. Thence, the MCNT-based LFI offers a rapid and low-cost tool for detecting CFB in human blood directly.
Collapse
|
19
|
Duan B, Hu X, Fan M, Xiong X, Han L, Wang Z, Tong D, Liu L, Wang X, Li W, Yang J, Huang C. RNA-Binding Motif Protein 6 is a Candidate Serum Biomarker for Pancreatic Cancer. Proteomics Clin Appl 2019; 13:e1900048. [PMID: 31207145 DOI: 10.1002/prca.201900048] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/26/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Early diagnosis is crucial to improve outcomes for pancreatic cancer patients (PC). The present study is designed to identify differently expressed peptides involved in PC as potential biomarkers. EXPERIMENTAL DESIGN The serum proteome of 22 PC patients, 12 pancreatitis patients (PP), and 45 healthy controls (HC) are analyzed using magnetic bead-based weak cation exchange (MB-WCX) and matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS). Next, a supervised neural network (SNN) algorithm model is established by ClinProTools and the candidate biomarker identified using liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS). Finally, the candidate biomarker is validated in tissue samples. RESULTS The SNN algorithm model discriminates PC from HC with 92.97% sensitivity and 94.55% specificity. Seventy-six differentially expressed peptides are identified, seven of which are significantly different among PC, PP, and HC (p < 0.05). Only one peak (m/z: 1466.99) tends to be upregulated in samples from HC, PP, and PC, which is identified as region of RNA-binding motif protein 6 (RBM6). In subsequent tissue analysis, it is verified that RBM6 expression is significantly higher in PC tissues than paracancerous tissue. CONCLUSIONS AND CLINICAL RELEVANCE The results indicate that RBM6 might serve as a candidate diagnostic biomarker for PC. CLINICAL RELEVANCE Methods used in this study could generate serum peptidome profiles of PC, PP, and HC, and present an approach to identify potential biomarkers for diagnosis of this malignancy.
Collapse
Affiliation(s)
- Baojun Duan
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China.,Department of Medical Oncology of Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Xiaoyan Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Meiyang Fan
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Xiaofan Xiong
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Lin Han
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Dongdong Tong
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Liying Liu
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Xiaofei Wang
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Wensheng Li
- Department of Pathology of Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Juan Yang
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Chen Huang
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| |
Collapse
|
20
|
Kim SH, Lee MJ, Hwang HK, Lee SH, Kim H, Paik YK, Kang CM. Prognostic potential of the preoperative plasma complement factor B in resected pancreatic cancer: A pilot study. Cancer Biomark 2019; 24:335-342. [DOI: 10.3233/cbm-181847] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Sung Hyun Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, College of Medicine, Yonsei University, Seoul, Korea
- Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Seoul, Korea
| | - Min Jung Lee
- Yonsei Proteome Research Center, Yonsei University, Seoul, Korea
- Department of Integrated OMICS for Biomedical Science, Yonsei University, Seoul, Korea
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Ho Kyung Hwang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, College of Medicine, Yonsei University, Seoul, Korea
- Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Seoul, Korea
| | - Sung Hwan Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, College of Medicine, Yonsei University, Seoul, Korea
- Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Seoul, Korea
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hoguen Kim
- Department of Pathology, College of Medicine, Yonsei University, Seoul, Korea
| | - Young-Ki Paik
- Yonsei Proteome Research Center, Yonsei University, Seoul, Korea
- Department of Integrated OMICS for Biomedical Science, Yonsei University, Seoul, Korea
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Chang Moo Kang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, College of Medicine, Yonsei University, Seoul, Korea
- Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Seoul, Korea
| |
Collapse
|
21
|
Costanza B, Rademaker G, Tiamiou A, De Tullio P, Leenders J, Blomme A, Bellier J, Bianchi E, Turtoi A, Delvenne P, Bellahcène A, Peulen O, Castronovo V. Transforming growth factor beta-induced, an extracellular matrix interacting protein, enhances glycolysis and promotes pancreatic cancer cell migration. Int J Cancer 2019; 145:1570-1584. [PMID: 30834519 DOI: 10.1002/ijc.32247] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 02/02/2019] [Accepted: 02/25/2019] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a deadly malignancy with no efficient therapy available up-to-date. Glycolysis is the main provider of energetic substrates to sustain cancer dissemination of PDAC. Accordingly, altering the glycolytic pathway is foreseen as a sound approach to trigger pancreatic cancer regression. Here, we show for the first time that high transforming growth factor beta-induced (TGFBI) expression in PDAC patients is associated with a poor outcome. We demonstrate that, although usually secreted by stromal cells, PDAC cells synthesize and secrete TGFBI in quantity correlated with their migratory capacity. Mechanistically, we show that TGFBI activates focal adhesion kinase signaling pathway through its binding to integrin αVβ5, leading to a significant enhancement of glycolysis and to the acquisition of an invasive phenotype. Finally, we show that TGFBI silencing significantly inhibits PDAC tumor development in a chick chorioallantoic membrane assay model. Our study highlights TGFBI as an oncogenic extracellular matrix interacting protein that bears the potential to serve as a target for new anti-PDAC therapeutic strategies.
Collapse
Affiliation(s)
- Brunella Costanza
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Gilles Rademaker
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Assia Tiamiou
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Pascal De Tullio
- Center for Interdisciplinary Research on Medicines, Metabolomics Group, University of Liège, Liège, Belgium
| | - Justine Leenders
- Center for Interdisciplinary Research on Medicines, Metabolomics Group, University of Liège, Liège, Belgium
| | - Arnaud Blomme
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Justine Bellier
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Elettra Bianchi
- Department of Pathology, University Hospital (CHU), University of Liège, Liège, Belgium
| | - Andrei Turtoi
- Tumor Microenvironment and Resistance to Treatment Laboratory, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
| | - Philippe Delvenne
- Department of Pathology, University Hospital (CHU), University of Liège, Liège, Belgium.,Laboratory of Experimental Pathology, GIGA Cancer, University of Liège, Liège, Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| |
Collapse
|
22
|
Follia L, Ferrero G, Mandili G, Beccuti M, Giordano D, Spadi R, Satolli MA, Evangelista A, Katayama H, Hong W, Momin AA, Capello M, Hanash SM, Novelli F, Cordero F. Integrative Analysis of Novel Metabolic Subtypes in Pancreatic Cancer Fosters New Prognostic Biomarkers. Front Oncol 2019; 9:115. [PMID: 30873387 PMCID: PMC6400843 DOI: 10.3389/fonc.2019.00115] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 02/07/2019] [Indexed: 12/25/2022] Open
Abstract
Background: Most of the patients with Pancreatic Ductal Adenocarcinoma (PDA) are not eligible for a curative surgical resection. For this reason there is an urgent need for personalized therapies. PDA is the result of complex interactions between tumor molecular profile and metabolites produced by its microenvironment. Despite recent studies identified PDA molecular subtypes, its metabolic classification is still lacking. Methods: We applied an integrative analysis on transcriptomic and genomic data of glycolytic genes in PDA. Data were collected from public datasets and molecular glycolytic subtypes were defined using hierarchical clustering. The grade of purity of the cancer samples was assessed estimating the different amount of stromal and immunological infiltrate among the identified PDA subtypes. Analyses of metabolomic data from a subset of PDA cell lines allowed us to identify the different metabolites produced by the metabolic subtypes. Sera of a cohort of 31 PDA patients were analyzed using Q-TOF mass spectrometer to measure the amount of metabolic circulating proteins present before and after chemotherapy. Results: Our integrative analysis of glycolytic genes identified two glycolytic and two non-glycolytic metabolic PDA subtypes. Glycolytic patients develop disease earlier, have poor prognosis, low immune-infiltrated tumors, and are characterized by a gain in chr12p13 genomic region. This gain results in the over-expression of GAPDH, TPI1, and FOXM1. PDA cell lines with the gain of chr12p13 are characterized by an higher lipid uptake and sensitivity to drug targeting the fatty acid metabolism. Our sera proteomic analysis confirms that TPI1 serum levels increase in poor prognosis gemcitabine-treated patients. Conclusions: We identify four metabolic PDA subtypes with different prognosis outcomes which may have pivotal role in setting personalized treatments. Moreover, our data suggest TPI1 as putative prognostic PDA biomarker.
Collapse
Affiliation(s)
- Laura Follia
- Center for Experimental Research and Medical Studies, Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Department of Computer Sciences, University of Turin, Turin, Italy
| | - Giulio Ferrero
- Department of Computer Sciences, University of Turin, Turin, Italy
| | - Giorgia Mandili
- Center for Experimental Research and Medical Studies, Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Marco Beccuti
- Department of Computer Sciences, University of Turin, Turin, Italy
| | - Daniele Giordano
- Center for Experimental Research and Medical Studies, Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
| | - Rosella Spadi
- Centro Oncologico Ematologico Subalpino, Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
| | - Maria Antonietta Satolli
- Centro Oncologico Ematologico Subalpino, Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
| | - Andrea Evangelista
- Servizio di Epidemiologia dei Tumori, Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
| | - Hiroyuki Katayama
- Department of Clinical Cancer Prevention Research, MD Anderson Cancer Center, Houston, TX, United States
| | - Wang Hong
- Department of Clinical Cancer Prevention Research, MD Anderson Cancer Center, Houston, TX, United States
| | - Amin A. Momin
- Department of Clinical Cancer Prevention Research, MD Anderson Cancer Center, Houston, TX, United States
| | - Michela Capello
- Department of Clinical Cancer Prevention Research, MD Anderson Cancer Center, Houston, TX, United States
| | - Samir M. Hanash
- Department of Clinical Cancer Prevention Research, MD Anderson Cancer Center, Houston, TX, United States
| | - Francesco Novelli
- Center for Experimental Research and Medical Studies, Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center, University of Turin, Turin, Italy
| | | |
Collapse
|
23
|
Dong D, Jia L, Zhang L, Ma N, Zhang A, Zhou Y, Ren L. Periostin and CA242 as potential diagnostic serum biomarkers complementing CA19.9 in detecting pancreatic cancer. Cancer Sci 2018; 109:2841-2851. [PMID: 29945294 PMCID: PMC6125476 DOI: 10.1111/cas.13712] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/24/2018] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor with few biomarkers to guide treatment options. Carbohydrate antigen 19.9 (CA19.9), the most frequently used biomarker for PDAC, is not sensitive and specific enough for the detection of the disease. This study aimed to evaluate serum periostin (POSTN) and CA242 as potential diagnostic biomarkers complementing CA19.9 in detecting pancreatic cancer. Blood samples were from 362 participants, including 213 patients with different stages of PDAC, 75 patients with benign pancreatic disease, and 74 healthy individuals. All samples were randomly divided into a training set and a validation set. Carbohydrate antigen 19.9, CA242, POSTN, as well as carcinoembryonic antigen, were measured by ELISA or automated immunoassay. The receiver operating characteristic curve analysis revealed that the performance of CA19.9 in the validation group were improved by the marker panel composed of CA19.9, POSTN, and CA242, to discriminate early stage PDAC not only from healthy controls (area under the curve [AUC]CA19.9 = 0.94 vs AUCCA19.9 + POSTN + CA242 = 0.98, P < .05) but also from benign conditions (AUCCA19.9 = 0.87 vs AUCCA19.9 + POSTN + CA242 = 0.90, P < .05). In addition, POSTN retained significant diagnostic capabilities to distinguish PDAC CA19.9-negative from healthy controls (AUCPOSTN = 0.87) as well as from benign conditions (AUCPOSTN = 0.84) in the whole set. This study suggested that POSTN and CA242 are potential diagnostic serum biomarkers complementing CA19.9 in detecting early pancreatic cancer.
Collapse
Affiliation(s)
- Dong Dong
- Department of LaboratoryTianjin Medical University Cancer Institute and HospitalTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjinChina
| | - Li Jia
- Department of LaboratoryTianjin Medical University Cancer Institute and HospitalTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjinChina
| | - Lufang Zhang
- Department of LaboratoryTianjin Medical University Cancer Institute and HospitalTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjinChina
| | - Na Ma
- Cancer BiobankTianjin Medical University Cancer Institute and HospitalTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjinChina
| | - Aimin Zhang
- Department of LaboratoryTianjin Medical University Cancer Institute and HospitalTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjinChina
| | - Yunli Zhou
- Department of LaboratoryTianjin Medical University Cancer Institute and HospitalTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjinChina
| | - Li Ren
- Department of LaboratoryTianjin Medical University Cancer Institute and HospitalTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjinChina
| |
Collapse
|
24
|
Long NP, Yoon SJ, Anh NH, Nghi TD, Lim DK, Hong YJ, Hong SS, Kwon SW. A systematic review on metabolomics-based diagnostic biomarker discovery and validation in pancreatic cancer. Metabolomics 2018; 14:109. [PMID: 30830397 DOI: 10.1007/s11306-018-1404-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/31/2018] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Metabolomics is an emerging approach for early detection of cancer. Along with the development of metabolomics, high-throughput technologies and statistical learning, the integration of multiple biomarkers has significantly improved clinical diagnosis and management for patients. OBJECTIVES In this study, we conducted a systematic review to examine recent advancements in the oncometabolomics-based diagnostic biomarker discovery and validation in pancreatic cancer. METHODS PubMed, Scopus, and Web of Science were searched for relevant studies published before September 2017. We examined the study designs, the metabolomics approaches, and the reporting methodological quality following PRISMA statement. RESULTS AND CONCLUSION: The included 25 studies primarily focused on the identification rather than the validation of predictive capacity of potential biomarkers. The sample size ranged from 10 to 8760. External validation of the biomarker panels was observed in nine studies. The diagnostic area under the curve ranged from 0.68 to 1.00 (sensitivity: 0.43-1.00, specificity: 0.73-1.00). The effects of patients' bio-parameters on metabolome alterations in a context-dependent manner have not been thoroughly elucidated. The most reported candidates were glutamic acid and histidine in seven studies, and glutamine and isoleucine in five studies, leading to the predominant enrichment of amino acid-related pathways. Notably, 46 metabolites were estimated in at least two studies. Specific challenges and potential pitfalls to provide better insights into future research directions were thoroughly discussed. Our investigation suggests that metabolomics is a robust approach that will improve the diagnostic assessment of pancreatic cancer. Further studies are warranted to validate their validity in multi-clinical settings.
Collapse
Affiliation(s)
- Nguyen Phuoc Long
- Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Sang Jun Yoon
- Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Nguyen Hoang Anh
- Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Tran Diem Nghi
- School of Medicine, Vietnam National University, Ho Chi Minh City, 700000, Vietnam
| | - Dong Kyu Lim
- Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Yu Jin Hong
- Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Soon-Sun Hong
- Department of Drug Development, College of Medicine, Inha University, Incheon, 22212, South Korea
| | - Sung Won Kwon
- Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
25
|
Irigoyen A, Jimenez-Luna C, Benavides M, Caba O, Gallego J, Ortuño FM, Guillen-Ponce C, Rojas I, Aranda E, Torres C, Prados J. Integrative multi-platform meta-analysis of gene expression profiles in pancreatic ductal adenocarcinoma patients for identifying novel diagnostic biomarkers. PLoS One 2018; 13:e0194844. [PMID: 29617451 PMCID: PMC5884535 DOI: 10.1371/journal.pone.0194844] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 03/09/2018] [Indexed: 01/16/2023] Open
Abstract
Applying differentially expressed genes (DEGs) to identify feasible biomarkers in diseases can be a hard task when working with heterogeneous datasets. Expression data are strongly influenced by technology, sample preparation processes, and/or labeling methods. The proliferation of different microarray platforms for measuring gene expression increases the need to develop models able to compare their results, especially when different technologies can lead to signal values that vary greatly. Integrative meta-analysis can significantly improve the reliability and robustness of DEG detection. The objective of this work was to develop an integrative approach for identifying potential cancer biomarkers by integrating gene expression data from two different platforms. Pancreatic ductal adenocarcinoma (PDAC), where there is an urgent need to find new biomarkers due its late diagnosis, is an ideal candidate for testing this technology. Expression data from two different datasets, namely Affymetrix and Illumina (18 and 36 PDAC patients, respectively), as well as from 18 healthy controls, was used for this study. A meta-analysis based on an empirical Bayesian methodology (ComBat) was then proposed to integrate these datasets. DEGs were finally identified from the integrated data by using the statistical programming language R. After our integrative meta-analysis, 5 genes were commonly identified within the individual analyses of the independent datasets. Also, 28 novel genes that were not reported by the individual analyses ('gained' genes) were also discovered. Several of these gained genes have been already related to other gastroenterological tumors. The proposed integrative meta-analysis has revealed novel DEGs that may play an important role in PDAC and could be potential biomarkers for diagnosing the disease.
Collapse
Affiliation(s)
- Antonio Irigoyen
- Department of Medical Oncology, Virgen de la Salud Hospital, Toledo, Spain
| | - Cristina Jimenez-Luna
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Manuel Benavides
- Department of Medical Oncology, Virgen de la Victoria Hospital, Malaga, Spain
| | - Octavio Caba
- Department of Health Sciences, University of Jaen, Jaen, Spain
| | - Javier Gallego
- Department of Medical Oncology, University General Hospital of Elche, Alicante, Spain
| | - Francisco Manuel Ortuño
- Department of Computer Architecture and Computer Technology, Research Center for Information and Communications Technologies, University of Granada, Granada, Spain
| | | | - Ignacio Rojas
- Department of Computer Architecture and Computer Technology, Research Center for Information and Communications Technologies, University of Granada, Granada, Spain
| | - Enrique Aranda
- Maimonides Institute of Biomedical Research (IMIBIC), Reina Sofia Hospital, University of Cordoba, Cordoba, Spain
| | - Carolina Torres
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
| |
Collapse
|
26
|
Jimenez-Luna C, Torres C, Ortiz R, Dieguez C, Martinez-Galan J, Melguizo C, Prados JC, Caba O. Proteomic biomarkers in body fluids associated with pancreatic cancer. Oncotarget 2018; 9:16573-16587. [PMID: 29662668 PMCID: PMC5893263 DOI: 10.18632/oncotarget.24654] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/25/2018] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PC) is a highly malignant disease that represents the fourth leading cancer-related death worldwide. There has been very little improvement in survival rates over recent years, and surgical resection remains the only reliable curative approach. Factors that contribute to this dismal prognosis for PC include its rapid progression and invasion, the absence of specific symptoms, and the little impact of available chemotherapy. Importantly, the management of this malignancy is also limited by the lack of highly specific and sensitive biomarkers for its diagnosis and follow-up, and their identification is therefore considered a promising strategy to improve outcomes in these patients. Numerous translational studies have explored the usefulness of body fluids as a non-invasive source of PC-specific biomarkers, and innovations in proteomic methods and technologies have provided a myriad of protein biomarkers for different cancers. The adoption of a proteomic approach has improved understanding of the biology of PC and contributed to the potential identification of protein biomarkers for this disease. This review considers the most recent research efforts to develop novel proteomic biomarkers in body fluids for PC.
Collapse
Affiliation(s)
- Cristina Jimenez-Luna
- Institute of Biopathology and Regenerative Medicine (IBIMER), Granada University, Granada, Spain
| | - Carolina Torres
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL, USA
| | - Raul Ortiz
- Department of Health Sciences, Jaen University, Jaen, Spain
| | - Carmelo Dieguez
- Department of Gastroenterology, San Cecilio University Hospital, Granada, Spain
| | | | - Consolacion Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Granada University, Granada, Spain
| | - Jose C. Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Granada University, Granada, Spain
| | - Octavio Caba
- Department of Health Sciences, Jaen University, Jaen, Spain
| |
Collapse
|
27
|
Wan W, Descalzo AB, Shinde S, Weißhoff H, Orellana G, Sellergren B, Rurack K. Ratiometric Fluorescence Detection of Phosphorylated Amino Acids Through Excited-State Proton Transfer by Using Molecularly Imprinted Polymer (MIP) Recognition Nanolayers. Chemistry 2017; 23:15974-15983. [PMID: 28869685 DOI: 10.1002/chem.201703041] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Indexed: 12/12/2022]
Abstract
A 2,3-diaminophenazine bis-urea fluorescent probe monomer (1) was developed. It responds to phenylphosphate and phosphorylated amino acids in a ratiometric fashion with enhanced fluorescence accompanied by the development of a redshifted emission band arising from an excited-state proton transfer (ESPT) process in the hydrogen-bonded probe/analyte complex. The two urea groups of 1 form a cleft-like binding pocket (Kb >1010 L2 mol-2 for 1:2 complex). Imprinting of 1 in presence of ethyl ester- and fluorenylmethyloxycarbonyl (Fmoc)-protected phosphorylated tyrosine (Fmoc-pTyr-OEt) as the template, methacrylamide as co-monomer, and ethyleneglycol dimethacrylate as cross-linker gave few-nanometer-thick molecularly imprinted polymer (MIP) shells on silica core microparticles with excellent selectivity for the template in a buffered biphasic assay. The supramolecular recognition features were established by spectroscopic and NMR studies. Rational screening of co-monomers and cross-linkers allowed to single out the best performing MIP components, giving significant imprinting factors (IF>3.5) while retaining ESPT emission and the ratiometric response in the thin polymer shell. Combination of the bead-based detection scheme with the phase-transfer assay dramatically improved the IF to 15.9, allowing sensitive determination of the analyte directly in aqueous media.
Collapse
Affiliation(s)
- Wei Wan
- Chemical and Optical Sensing Division, Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Strasse 11, 12489, Berlin, Germany
| | - Ana B Descalzo
- Department of Organic Chemistry, Complutense University of Madrid (UCM), 28040, Madrid, Spain
| | - Sudhirkumar Shinde
- Department of Biomedical Science, Malmö University, 20506, Malmö, Sweden
| | - Hardy Weißhoff
- Department of Chemistry, Humboldt University Berlin, Brook-Taylor-Strasse 2, 12489, Berlin, Germany
| | - Guillermo Orellana
- Department of Organic Chemistry, Complutense University of Madrid (UCM), 28040, Madrid, Spain
| | - Börje Sellergren
- Department of Biomedical Science, Malmö University, 20506, Malmö, Sweden
| | - Knut Rurack
- Chemical and Optical Sensing Division, Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Strasse 11, 12489, Berlin, Germany
| |
Collapse
|
28
|
Nedaeinia R, Manian M, Jazayeri MH, Ranjbar M, Salehi R, Sharifi M, Mohaghegh F, Goli M, Jahednia SH, Avan A, Ghayour-Mobarhan M. Circulating exosomes and exosomal microRNAs as biomarkers in gastrointestinal cancer. Cancer Gene Ther 2017; 24:48-56. [PMID: 27982021 DOI: 10.1038/cgt.2016.77] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/13/2016] [Accepted: 11/14/2016] [Indexed: 02/07/2023]
Abstract
The most important biological function of exosomes is their possible use as biomarkers in clinical diagnosis. Compared with biomarkers identified in conventional specimens such as serum or urine, exosomal biomarkers provide the highest amount of sensitivity and specificity, which can be attributed to their excellent stability. Exosomes, which harbor different types of proteins, nucleic acids and lipids, are present in almost all bodily fluids. The molecular constituents of exosomes, especially exosomal proteins and microRNAs (miRNAs), are promising as biomarkers in clinical diagnosis. This discovery that exosomes also contain messenger RNAs and miRNAs shows that they could be carriers of genetic information. Although the majority of RNAs found in exosomes are degraded RNA fragments with a length of <200 nucleotides, some full-length RNAs might be present that may affect protein production in the recipient cell. In addition, exosomal miRNAs have been found to be associated with certain diseases. Several studies have pointed out miRNA contents of circulating exosomes that are similar to those of originating cancer cells. In this review, the recent advances in circulating exosomal miRNAs as biomarkers in gastrointestinal cancers are discussed. These studies indicated that miRNAs can be detected in exosomes isolated from body fluids such as saliva, which suggests potential advantages of using exosomal miRNAs as noninvasive novel biomarkers.
Collapse
Affiliation(s)
- R Nedaeinia
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology, Student Research Committee, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - M Manian
- Immunology Research Center, Department of Immunology, Iran University of Medical Science, Tehran, Iran
| | - M H Jazayeri
- Immunology Research Center, Department of Immunology, Iran University of Medical Science, Tehran, Iran
| | - M Ranjbar
- Deputy of Food and Drug, Department of Food Control Administration, Isfahan University of Medical Sciences, Isfahan, Iran
| | - R Salehi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - M Sharifi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - F Mohaghegh
- Department of Radiotherapy, Arak University of Medical Sciences, Arak, Iran
| | - M Goli
- Department of Food Science and Technology, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| | - S H Jahednia
- Department of Immunology, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - A Avan
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - M Ghayour-Mobarhan
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Biochemistry of Nutrition Research Center, Department of Nutrition, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
29
|
Halbrook CJ, Lyssiotis CA. Employing Metabolism to Improve the Diagnosis and Treatment of Pancreatic Cancer. Cancer Cell 2017; 31:5-19. [PMID: 28073003 DOI: 10.1016/j.ccell.2016.12.006] [Citation(s) in RCA: 278] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/03/2016] [Accepted: 12/14/2016] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma is on pace to become the second leading cause of cancer-related death. The high mortality rate results from a lack of methods for early detection and the inability to successfully treat patients once diagnosed. Pancreatic cancer cells have extensively reprogrammed metabolism, which is driven by oncogene-mediated cell-autonomous pathways, the unique physiology of the tumor microenvironment, and interactions with non-cancer cells. In this review, we discuss how recent efforts delineating rewired metabolic networks in pancreatic cancer have revealed new in-roads to develop detection and treatment strategies for this dreadful disease.
Collapse
Affiliation(s)
- Christopher J Halbrook
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
30
|
Guo D, Guo J, Yao J, Jiang K, Hu J, Wang B, Liu H, Lin L, Sun W, Jiang X. D-dopachrome tautomerase is over-expressed in pancreatic ductal adenocarcinoma and acts cooperatively with macrophage migration inhibitory factor to promote cancer growth. Int J Cancer 2016; 139:2056-67. [PMID: 27434219 DOI: 10.1002/ijc.30278] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 05/25/2016] [Accepted: 07/12/2016] [Indexed: 12/18/2022]
Abstract
Previous studies have established the important role of MIF in the development of pancreatic ductal adenocarcinoma (PDAC) for both therapeutic and diagnostic perspectives, but little is known about the expression and function of D-dopachrome tautomerase (DDT), a functional homolog of MIF, in PDAC. In the present study, we demonstrated that DDT was over-expressed in PDAC tissues in a pattern correlated with MIF. In the pancreatic cancer cell lines, PANC-1, BXPC-3 and ASPC-1, both DDT and MIF were expressed and co-localized with each other in the endosomal compartments and plasma membrane. Knockdown of DDT and MIF in PANC-1 cells cooperatively inhibited ERK1/2 and AKT phosphorylation, increased p53 expression, and reduced cell proliferation, invasion and tumor formation. These effects were rescued by the re-expression of MIF or DDT, but not by the forced expression of the tautomerase-deficient mutants of DDT and MIF, P1G-DDT and P1G-MIF. Finally, we observed that 4-iodo-6-phenylpyrimidine (4-IPP), a covalent tautomerase inhibitor of both DDT and MIF, attenuated PANC-1 cell proliferation and colony formation in vitro and tumor growth in vivo. Thus, targeting the tautomerase sites of both MIF and DDT may offer more efficient therapeutic benefits to PDAC patients.
Collapse
Affiliation(s)
- Dawei Guo
- Department of Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Jinshuai Guo
- Department of Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Junchao Yao
- Department of Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Kun Jiang
- Department of Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Jianhua Hu
- Department of Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bo Wang
- Department of Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Haiyang Liu
- Department of Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Lin Lin
- Department of Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Wenyu Sun
- Department of Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaofeng Jiang
- Department of Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
31
|
Long J, Liu Z, Wu X, Xu Y, Ge C. Screening for genes and subnetworks associated with pancreatic cancer based on the gene expression profile. Mol Med Rep 2016; 13:3779-86. [PMID: 27035224 PMCID: PMC4838159 DOI: 10.3892/mmr.2016.5007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 02/17/2016] [Indexed: 12/27/2022] Open
Abstract
The present study aimed to screen for potential genes and subnetworks associated with pancreatic cancer (PC) using the gene expression profile. The expression profile GSE 16515 was downloaded from the Gene Expression Omnibus database, which included 36 PC tissue samples and 16 normal samples. Limma package in R language was used to screen differentially expressed genes (DEGs), which were grouped as up‑ and downregulated genes. Then, PFSNet was applied to perform subnetwork analysis for all the DEGs. Moreover, Gene Ontology (GO) and REACTOME pathway enrichment analysis of up‑ and downregulated genes was performed, followed by protein‑protein interaction (PPI) network construction using Search Tool for the Retrieval of Interacting Genes Search Tool for the Retrieval of Interacting Genes. In total, 1,989 DEGs including 1,461 up‑ and 528 downregulated genes were screened out. Subnetworks including pancreatic cancer in PC tissue samples and intercellular adhesion in normal samples were identified, respectively. A total of 8 significant REACTOME pathways for upregulated DEGs, such as hemostasis and cell cycle, mitotic were identified. Moreover, 4 significant REACTOME pathways for downregulated DEGs, including regulation of β‑cell development and transmembrane transport of small molecules were screened out. Additionally, DEGs with high connectivity degrees, such as CCNA2 (cyclin A2) and PBK (PDZ binding kinase), of the module in the protein‑protein interaction network were mainly enriched with cell‑division cycle. CCNA2 and PBK of the module and their relative pathway cell‑division cycle, and two subnetworks (pancreatic cancer and intercellular adhesion subnetworks) may be pivotal for further understanding of the molecular mechanism of PC.
Collapse
Affiliation(s)
- Jin Long
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhe Liu
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xingda Wu
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuanhong Xu
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Chunlin Ge
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
32
|
Liu P, Zhu Y, Liu L. Elevated serum CA72-4 levels predict poor prognosis in pancreatic adenocarcinoma after intensity-modulated radiation therapy. Oncotarget 2016; 6:9592-9. [PMID: 25860937 PMCID: PMC4496241 DOI: 10.18632/oncotarget.3562] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 02/10/2015] [Indexed: 12/20/2022] Open
Abstract
Carbohydrate antigen 72-4 (CA72-4) is a human tumor-associated glycoprotein, commonly used as a tumor marker for diagnosing and predicting outcome in gastric and ovarian cancers. However, the relationship between serum CA72-4 levels and prognosis of pancreatic adenocarcinoma has not been fully elucidated. A total of 113 consecutive locally advanced pancreatic adenocarcinoma patients who underwent intensity-modulated radiation therapy (IMRT) with or without chemotherapy were enrolled in this study. Serum CA72-4 levels were analyzed using immunoenzymometric assays. The association between serum CA72-4 levels and prognosis was evaluated. Serum CA72-4 levels was related with lymph node metastasis (P<0.001). The median overall survival time was 14.0 months for patients with serum CA72-4 normal levels and 10.0 months for the elevated levels (P<0.001). Multivariate analysis identified that Serum CA72-4 concentration was a significant prognostic factor (P<0.001). The hazard ratio (HR) of elevated serum CA72-4 levels compared with normal serum CA72-4 levels was 2.34 (95% confidence interval [CI]: 1.46-3.73), after adjusted for gender and age. Based on this finding, Serum CA72-4 is a potential marker to predict lymph node metastasis and prognosis in pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Peng Liu
- Department of Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yuan Zhu
- Department of Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Luying Liu
- Department of Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
33
|
Kang M, Qin W, Buya M, Dong X, Zheng W, Lu W, Chen J, Guo Q, Wu Y. VNN1, a potential biomarker for pancreatic cancer-associated new-onset diabetes, aggravates paraneoplastic islet dysfunction by increasing oxidative stress. Cancer Lett 2016; 373:241-50. [PMID: 26845448 DOI: 10.1016/j.canlet.2015.12.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 12/11/2015] [Accepted: 12/25/2015] [Indexed: 12/28/2022]
Abstract
In our previous clinical microarray analysis, we were the first to report on Vanin-1 (VNN1) as a novel clinically derived biomarker of pancreatic cancer-associated new-onset diabetes (PCAND). The functional mechanisms of VNN1 in the pathogenesis of PCAND, however, are not completely understood. In the present study, we further extend our previous clinical study to include laboratory research. The functions and mechanisms of neoplastic overexpressed VNN1 in PCAND have been explored using a co-culture model. Furthermore, the serum concentrations and discrimination power of downstream molecules of VNN1 were tested in a PCAND cohort. Pancreatic ductal adenocarcinoma (PDA) overexpressed VNN1 further aggravates paraneoplastic islet dysfunction; decreases in GSH/PPAR-γ concentrations and increases in ROS/cysteamine might be primary cause of this effect. Clinical serum analyses revealed that the expression profiles of these molecules were aberrant in the PCAND group. Our results further demonstrated that PCAND is a type of paraneoplastic diabetes. As the only clinically derived biomarker for PCAND screening available today, the biological role of VNN1 in triggering oxidative stress within the pancreatic microenvironment is important. The molecules downstream of VNN1 are also potential biomarkers for PCAND screening.
Collapse
Affiliation(s)
- Muxing Kang
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China; Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Wenjie Qin
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China; Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Miranbieke Buya
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Xin Dong
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Wen Zheng
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Wenjie Lu
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Jian Chen
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Qingqu Guo
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Yulian Wu
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China; Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China.
| |
Collapse
|
34
|
YU XINZHE, LI HENGCHAO, FU DELIANG, JIN CHEN, LI JI. Characterization of the role of the photosensitizer, deuteporfin, in the detection of lymphatic metastases in a pancreatic cancer xenograft model. Oncol Lett 2015; 10:1430-1436. [PMID: 26622685 PMCID: PMC4533294 DOI: 10.3892/ol.2015.3441] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 06/05/2015] [Indexed: 12/31/2022] Open
Abstract
Currently, the use of photosensitizers as tracer agents to detect lymphatic metastases is a developing area of study in the field of pancreatic cancer treatment. In the present study, deuteporfin, a novel photosensitizer, was used as a tracer agent to detect lymphatic metastases in a pancreatic cancer xenograft model. The biodistribution and pharmacokinetics of deuteporfin, following intravenous administration and injection of deuteporfin into the left rear footpad, were investigated in Sprague-Dawley rats. The increased difference in deuteporfin concentration between the cancerous and normal tissues was directly observed through the application of a Wood's lamp. In addition, the highly lymphatic BxPC-3-LN5 human metastatic pancreatic cancer cell line was generated from BxPC-3 cells using a continuous screening and seeding method in vivo. A xenograft model of the BxPC-3-LN5 human pancreatic cancer cell line transplanted into the left rear footpad of nude mice, was established. The effects of deuteporfin as a tracer agent in the detection of lymphatic metastases were then characterized in the pancreatic cancer xenograft model. Following intravenous administration, deuteporfin was rapidly enriched in the pancreas and popliteal fossa lymph nodes compared with that of the left rear footpad administration group. In addition, deuteporfin appeared to be selectively enriched in the cancerous pancreatic lymph nodes of the pancreatic cancer xenograft model. These results indicated that deuteporfin may be developed as a novel photosensitizer tracer agent for the detection of lymphatic metastases in pancreatic cancer. The advantages of deuteporfin are that it has a selective tumor-targeting effect due to high tissue uptake, and that it may be administered intravenously and is therefore suitable for surgery.
Collapse
Affiliation(s)
- XINZHE YU
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - HENGCHAO LI
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - DELIANG FU
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - CHEN JIN
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - JI LI
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
35
|
Kwon MS, Kim Y, Lee S, Namkung J, Yun T, Yi SG, Han S, Kang M, Kim SW, Jang JY, Park T. Integrative analysis of multi-omics data for identifying multi-markers for diagnosing pancreatic cancer. BMC Genomics 2015; 16 Suppl 9:S4. [PMID: 26328610 PMCID: PMC4547403 DOI: 10.1186/1471-2164-16-s9-s4] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background microRNA (miRNA) expression plays an influential role in cancer classification and malignancy, and miRNAs are feasible as alternative diagnostic markers for pancreatic cancer, a highly aggressive neoplasm with silent early symptoms, high metastatic potential, and resistance to conventional therapies. Methods In this study, we evaluated the benefits of multi-omics data analysis by integrating miRNA and mRNA expression data in pancreatic cancer. Using support vector machine (SVM) modelling and leave-one-out cross validation (LOOCV), we evaluated the diagnostic performance of single- or multi-markers based on miRNA and mRNA expression profiles from 104 PDAC tissues and 17 benign pancreatic tissues. For selecting even more reliable and robust markers, we performed validation by independent datasets from the Gene Expression Omnibus (GEO) and the Cancer Genome Atlas (TCGA) data depositories. For validation, miRNA activity was estimated by miRNA-target gene interaction and mRNA expression datasets in pancreatic cancer. Results Using a comprehensive identification approach, we successfully identified 705 multi-markers having powerful diagnostic performance for PDAC. In addition, these marker candidates annotated with cancer pathways using gene ontology analysis. Conclusions Our prediction models have strong potential for the diagnosis of pancreatic cancer.
Collapse
|
36
|
Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol 2015; 17:816-26. [PMID: 25985394 DOI: 10.1038/ncb3169] [Citation(s) in RCA: 2028] [Impact Index Per Article: 202.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 03/26/2015] [Indexed: 11/09/2022]
Abstract
Pancreatic ductal adenocarcinomas (PDACs) are highly metastatic with poor prognosis, mainly due to delayed detection. We hypothesized that intercellular communication is critical for metastatic progression. Here, we show that PDAC-derived exosomes induce liver pre-metastatic niche formation in naive mice and consequently increase liver metastatic burden. Uptake of PDAC-derived exosomes by Kupffer cells caused transforming growth factor β secretion and upregulation of fibronectin production by hepatic stellate cells. This fibrotic microenvironment enhanced recruitment of bone marrow-derived macrophages. We found that macrophage migration inhibitory factor (MIF) was highly expressed in PDAC-derived exosomes, and its blockade prevented liver pre-metastatic niche formation and metastasis. Compared with patients whose pancreatic tumours did not progress, MIF was markedly higher in exosomes from stage I PDAC patients who later developed liver metastasis. These findings suggest that exosomal MIF primes the liver for metastasis and may be a prognostic marker for the development of PDAC liver metastasis.
Collapse
|
37
|
Roesli C. Recent advances in proteomically subtyping pancreatic ductal adenocarcinomas and their potential clinical impact. Expert Rev Proteomics 2015; 12:5-8. [PMID: 25407217 DOI: 10.1586/14789450.2015.983478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with a median overall survival of 6 months. Late diagnosis due to the absence of specific symptoms during disease development, in addition to extensive metastatic potential and resistance to chemotherapy and radiotherapy, are the most important reasons for short survival. Research efforts have therefore been focused on the development of early disease detection. However, the only US FDA-approved clinical biomarker, CA19-9, is considered inapplicable for screening and/or early detection of PDAC. The following editorial provides the reader with a short introduction to the topic of PDAC and gives focus to the current state of proteomic research in the field of PDAC biomarker discovery. This editorial also highlights the efforts made to subdivide this tumor entity and the potential clinical impact of patient stratification. Finally, the author provides opinions on the impact of proteomics to PDAC subtype stratification over the next 5 years.
Collapse
Affiliation(s)
- Christoph Roesli
- Junior Research Group Biomarker Discovery, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
38
|
Chan A, Prassas I, Dimitromanolakis A, Brand RE, Serra S, Diamandis EP, Blasutig IM. Validation of biomarkers that complement CA19.9 in detecting early pancreatic cancer. Clin Cancer Res 2014; 20:5787-95. [PMID: 25239611 PMCID: PMC4233184 DOI: 10.1158/1078-0432.ccr-14-0289] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is a significant cause of cancer mortality. Carbohydrate antigen 19.9 (CA19.9), the only tumor marker available to detect and monitor PDAC, is not sufficiently sensitive and specific to consistently differentiate early cancer from benign disease. In this study, we aimed to validate recently discovered serum protein biomarkers for the early detection of PDAC and ultimately develop a biomarker panel that could discriminate PDAC from other benign disease better than the existing marker CA19.9. PATIENTS AND METHODS We performed a retrospective blinded evaluation of 400 serum samples collected from individuals recruited on a consecutive basis. The sample population consisted of 250 individuals with PDAC at various stages, 130 individuals with benign conditions and 20 healthy individuals. The serum levels of each biomarker were determined by ELISAs or automated immunoassay. RESULTS By randomly splitting matched samples into a training (n = 186) and validation (n = 214) set, we were able to develop and validate a biomarker panel consisting of CA19.9, CA125, and LAMC2 that significantly improved the performance of CA19.9 alone. Improved discrimination was observed in the validation set between all PDAC and benign conditions (AUCCA19.9 = 0.80 vs. AUCCA19.9+CA125+LAMC2 = 0.87; P < 0.005) as well as between early-stage PDAC and benign conditions (AUCCA19.9 = 0.69 vs. AUCCA19.9+CA125+LAMC2 = 0.76; P < 0.05) and between early-stage PDAC and chronic pancreatitis (CP; AUCCA19.9 = 0.59 vs. AUCCA19.9+CA125+LAMC2 = 0.74; P < 0.05). CONCLUSIONS The data demonstrate that a serum protein biomarker panel consisting of CA125, CA19.9, and LAMC2 is able to significantly improve upon the performance of CA19.9 alone in detecting PDAC.
Collapse
Affiliation(s)
- Alison Chan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada. Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Ioannis Prassas
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada. Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | | | - Randall E Brand
- University of Pittsburgh, Division of Gastroenterology, Hepatology & Nutrition, Pittsburgh, Pennsylvania
| | - Stefano Serra
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada. Department of Pathology, University Health Network, Toronto, Ontario, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada. Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada. Department of Clinical Biochemistry, University Health Network, Toronto, Ontario, Canada
| | - Ivan M Blasutig
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada. Department of Clinical Biochemistry, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
39
|
Kwon MS, Kim Y, Lee S, Namkung J, Yun T, Yi SG, Han S, Kang M, Kim SW, Jang JY, Park T. Biomarker development for pancreatic ductal adenocarcinoma using integrated analysis of mRNA and miRNA expression. 2014 IEEE INTERNATIONAL CONFERENCE ON BIOINFORMATICS AND BIOMEDICINE (BIBM) 2014:273-278. [DOI: 10.1109/bibm.2014.6999167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
40
|
Pysz MA, Machtaler SB, Seeley ES, Lee JJ, Brentnall TA, Rosenberg J, Tranquart F, Willmann JK. Vascular endothelial growth factor receptor type 2-targeted contrast-enhanced US of pancreatic cancer neovasculature in a genetically engineered mouse model: potential for earlier detection. Radiology 2014; 274:790-9. [PMID: 25322341 DOI: 10.1148/radiol.14140568] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE To test ultrasonographic (US) imaging with vascular endothelial growth factor receptor type 2 (VEGFR2)-targeted microbubble contrast material for the detection of pancreatic ductal adenocarcinoma (PDAC) in a transgenic mouse model of pancreatic cancer development. MATERIALS AND METHODS Experiments involving animals were approved by the Institutional Administrative Panel on Laboratory Animal Care at Stanford University. Transgenic mice (n = 44; Pdx1-Cre, KRas(G12D), Ink4a(-/-)) that spontaneously develop PDAC starting at 4 weeks of age were imaged by using a dedicated small-animal US system after intravenous injection of 5 × 10(7) clinical-grade VEGFR2-targeted microbubble contrast material. The pancreata in wild-type (WT) mice (n = 64) were scanned as controls. Pancreatic tissue was analyzed ex vivo by means of histologic examination (with hematoxylin-eosin staining) and immunostaining of vascular endothelial cell marker CD31 and VEGFR2. The Wilcoxon rank sum test and linear mixed-effects model were used for statistical analysis. RESULTS VEGFR2-targeted US of PDAC showed significantly higher signal intensities (26.8-fold higher; mean intensity ± standard deviation, 6.7 linear arbitrary units [lau] ± 8.5; P < .001) in transgenic mice compared with normal, control pancreata of WT mice (mean intensity, 0.25 lau ± 0.25). The highest VEGFR2-targeted US signal intensities were observed in smaller tumors, less than 3 mm in diameter (30.8-fold higher than control tissue with mean intensity of 7.7 lau ± 9.3 [P < .001]; and 1.7-fold higher than lesions larger than 3 mm in diameter with mean intensity of 4.6 lau ± 5.8 [P < .024]). Ex vivo quantitative VEGFR2 immunofluorescence demonstrated that VEGFR2 expression was significantly higher in pancreatic tumors (P < .001; mean fluorescent intensity, 499.4 arbitrary units [au] ± 179.1) compared with normal pancreas (mean fluorescent intensity, 232.9 au ± 83.7). CONCLUSION US with clinical-grade VEGFR2-targeted microbubbles allows detection of small foci of PDAC in transgenic mice.
Collapse
Affiliation(s)
- Marybeth A Pysz
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford University, 300 Pasteur Dr, Room H1307, Stanford, CA 94305 (M.A.P., S.B.M., J.R., J.K.W.); Department of Pathology, University of California at San Francisco, San Francisco, Calif (E.S.S.); Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Howard Hughes Medical Institute, Stanford School of Medicine, Stanford University, Stanford, Calif (J.J.L.); Department of Medicine, University of Washington, Seattle, Wash (T.A.B.); and Bracco Suisse SA, Geneva, Switzerland (F.T.)
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Torres C, Perales S, Alejandre MJ, Iglesias J, Palomino RJ, Martin M, Caba O, Prados JC, Aránega A, Delgado JR, Irigoyen A, Ortuño FM, Rojas I, Linares A. Serum cytokine profile in patients with pancreatic cancer. Pancreas 2014; 43:1042-1049. [PMID: 24979617 DOI: 10.1097/mpa.0000000000000155] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma is a deadly disease because of late diagnosis and chemoresistance. We aimed to find a panel of serum cytokines representing diagnostic and predictive biomarkers for pancreatic cancer. METHODS A cytokine antibody array was performed to simultaneously identify 507 cytokines in sera of patients with pancreatic cancer and healthy controls. The nonparametric Mann-Whitney U test was used to pairwise compare the controls, the pretreated patients, and the posttreated patients. Fold changes greater than or equal to 1.5 or less than or equal to 1/1.5 were considered significant. Receiver operating characteristic curves were used to assess the performance of the model. A leave-one-out cross-validation was used for estimating prediction error. RESULTS Comparing the sera of pretreated patients against the control samples, the cytokines fibroblast growth factor 10 (FGF-10/keratinocyte growth factor-2 (KGF-2), chemokine (C-X-C motif) ligand 11 interferon inducible T cell alpha chemokine (I-TAC)/chemokine [C-X-C motif] ligand 11 (CXCL11), oncostatin M (OSM), osteoactivin/glycoprotein nonmetastatic melanoma protein B, and stem cell factor (SCF) were found significantly overexpressed. Besides, the cytokines CD30 ligand/tumor necrosis factor superfamily, member 8 (TNFSF8), chordin-like 2, FGF-10/KGF-2, growth/differentiation factor 15, I-TAC/CXCL11, OSM, and SCF were differentially expressed in response to treatment. CONCLUSIONS We propose a role for FGF-10/KGF-2, I-TAC/CXCL11, OSM, osteoactivin/glycoprotein nonmetastatic melanoma protein B, and SCF as novel diagnostic biomarkers. CD30 ligand/TNFSF8, chordin-like 2, FGF-10/KGF-2, growth/differentiation factor 15, I-TAC/CXCL11, OSM, and SCF might represent as predictive biomarkers for gemcitabine and erlotinib response of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Carolina Torres
- From the *Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada; †Department of Molecular Oncology, Pfizer-University of Granada Center for Genomics and Oncological Research, Granada; ‡Department of Health Sciences, University of Jaen, Jaen; §Department of Human Anatomy and Embryology, University of Granada; ∥Oncology Service, Virgen de las Nieves Hospital; and ¶Department of Computer Architecture and Computer Technology, University of Granada, Granada, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lee MJ, Na K, Jeong SK, Lim JS, Kim SA, Lee MJ, Song SY, Kim H, Hancock WS, Paik YK. Identification of human complement factor B as a novel biomarker candidate for pancreatic ductal adenocarcinoma. J Proteome Res 2014; 13:4878-88. [PMID: 25057901 DOI: 10.1021/pr5002719] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pancreatic cancer (PC; pancreatic ductal adenocarcinoma) is characterized by significant morbidity and mortality worldwide. Although carbohydrate antigen (CA) 19-9 has been known as a PC biomarker, it is not commonly used for general screening because of its low sensitivity and specificity. Therefore, there is an urgent need to develop a new biomarker for PC diagnosis in the earlier stage of cancer. To search for a novel serologic PC biomarker, we carried out an integrated proteomic analysis for a total of 185 pooled or individual plasma from healthy donors and patients with five disease groups including chronic pancreatitis (CP), PC, and other cancers (e.g., hepatocellular carcinoma, cholangiocarcinoma, and gastric cancer) and identified complement factor b (CFB) as a candidate serologic biomarker for PC diagnosis. Immunoblot analysis of CFB revealed more than two times higher expression in plasma samples from PC patients compared with plasma from individuals without PC. Immunoprecipitation coupled to mass spectrometry analysis confirmed both molecular identity and higher expression of CFB in PC samples. CFB showed distinctly higher specificity than CA 19-9 for PC against other types of digestive cancers and in discriminating PC patients from non-PC patients (p < 0.0001). In receiver operator characteristic curve analysis, CFB showed an area under curve of 0.958 (95% CI: 0.956 to 0.959) compared with 0.833 (95% CI: 0.829 to 0.837) for CA 19-9. Furthermore, the Y-index of CFB was much higher than that of CA 19-9 (71.0 vs 50.4), suggesting that CFB outperforms CA 19-9 in discriminating PC from CP and other gastrointestinal cancers. This was further supported by immunoprecipitation and qRT-PCR assays showing higher expression of CFB in PC cell lines than in normal cell lines. A combination of CFB and CA 19-9 showed markedly improved sensitivity (90.1 vs 73.1%) over that of CFB alone in the diagnosis of PC against non-PC, with similar specificity (97.2 vs 97.9%). Thus, our results identify CFB as a novel serologic PC biomarker candidate and warrant further investigation into a large-scale validation and its role in molecular mechanism of pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Min Jung Lee
- Yonsei Proteome Research Center and ‡Department of Integrated OMICS for Biomedical Science and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University , 50 Yonsei-ro, Sudaemoon-ku, Seoul 120-749, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Oberg HH, Peipp M, Kellner C, Sebens S, Krause S, Petrick D, Adam-Klages S, Röcken C, Becker T, Vogel I, Weisner D, Freitag-Wolf S, Gramatzki M, Kabelitz D, Wesch D. Novel bispecific antibodies increase γδ T-cell cytotoxicity against pancreatic cancer cells. Cancer Res 2014; 74:1349-60. [PMID: 24448235 DOI: 10.1158/0008-5472.can-13-0675] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The ability of human γδ T cells from healthy donors to kill pancreatic ductal adenocarcinoma (PDAC) in vitro and in vivo in immunocompromised mice requires the addition of γδ T-cell-stimulating antigens. In this study, we demonstrate that γδ T cells isolated from patients with PDAC tumor infiltrates lyse pancreatic tumor cells after selective stimulation with phosphorylated antigens. We determined the absolute numbers of γδ T-cell subsets in patient whole blood and applied a real-time cell analyzer to measure their cytotoxic effector function over prolonged time periods. Because phosphorylated antigens did not optimally enhance γδ T-cell cytotoxicity, we designed bispecific antibodies that bind CD3 or Vγ9 on γδ T cells and Her2/neu (ERBB2) expressed by pancreatic tumor cells. Both antibodies enhanced γδ T-cell cytotoxicity with the Her2/Vγ9 antibody also selectively enhancing release of granzyme B and perforin. Supporting these observations, adoptive transfer of γδ T cells with the Her2/Vγ9 antibody reduced growth of pancreatic tumors grafted into SCID-Beige immunocompromised mice. Taken together, our results show how bispecific antibodies that selectively recruit γδ T cells to tumor antigens expressed by cancer cells illustrate the tractable use of endogenous γδ T cells for immunotherapy.
Collapse
Affiliation(s)
- Hans-Heinrich Oberg
- Authors' Affiliations: Institute of Immunology; Division of Stem Cell Transplantation and Immunotherapy; Institute for Experimental Medicine; Institute of Pathology; Clinic of General and Thoracic Surgery; Institute for Medical Informatics and Statistic, Christian-Albrechts-University Kiel; Municipal Hospital, Department of Surgery; and Clinic of Gynaecology and Obstetrics, Kiel, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ballehaninna UK, Chamberlain RS. Biomarkers for pancreatic cancer: promising new markers and options beyond CA 19-9. Tumour Biol 2013; 34:3279-92. [PMID: 23949878 DOI: 10.1007/s13277-013-1033-3] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 07/18/2013] [Indexed: 12/11/2022] Open
Abstract
Pancreatic adenocarcinoma accounts for nearly 90-95% of exocrine malignant tumors of the pancreas. Traditionally, overexpressed proteins/epitopes such as CA 19-9, CA-50, CEA, and many others were being used as pancreatic cancer tumor markers. The main utility of these biomarkers was in the diagnosis of pancreatic cancer as well as to assess response to chemotherapy and to determine prognosis and to predict tumor recurrence. However, these markers had significant limitations such as lack of sensitivity, false-negative results in certain blood groups, as well as false-positive elevation in the presence of obstructive jaundice. To circumvent these limitations, an extraordinary amount of research is being performed to identify an accurate tumor marker or a panel of markers that could aid in the management of the pancreatic cancer. Although this research has identified a large number and different variety of biomarkers, few hold future promise as a preferred marker for pancreatic cancer. This review provides an insight into exciting new areas of pancreatic biomarker research such as salivary, pancreatic juice, and stool markers that can be used as a noninvasive test to identify pancreatic cancer. This manuscript also provides a discussion on newer biomarkers, the role of microRNAs, and pancreatic cancer proteomics, which have the potential to identify a preferred tumor marker for pancreatic adenocarcinoma. This review further elaborates on important genetic changes associated with the development and progression of pancreatic cancer that holds the key for the identification of a sensitive biomarker and which could also serve as a therapeutic target.
Collapse
Affiliation(s)
- Umashankar K Ballehaninna
- Department of Surgery, Saint Barnabas Medical Center, 94, Old Short Hills Road, Livingston, NJ, 07039, USA
| | | |
Collapse
|