1
|
Qiao Z, Wang E, Bao B, Tan X, Chen H, Wang D, Yuan L. Diagnostic and prognostic value of circulating exosomal glypican-1 in pancreatic cancer: a meta-analysis. Lab Med 2024; 55:543-552. [PMID: 38470244 DOI: 10.1093/labmed/lmae013] [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] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is usually detected in the advanced stages. Liquid biopsy has become a revolutionary strategy for cancer diagnosis and prognosis prediction. This study aims to investigate the diagnostic and prognostic value of circulating exosomal glypican-1 (GPC-1) in PC. METHODS We systematically searched relevant studies. For diagnostic accuracy, pooled sensitivity and specificity and the area under the summary receiver operating characteristic curve (AUC) were calculated. Regarding prognostic value, hazard ratios (HRs) and 95% CIs for overall survival (OS) were summarized by using a random-effects model. RESULTS We found 8 studies that examined the diagnostic value of circulating exosomal GPC-1 in PC, and 3 studies that investigated its prognostic value. Pooled sensitivity and specificity were 0.88 (95% CI, 0.65-0.97) and 0.86 (95% CI, 0.72-0.94). The AUC was 0.93 (95% CI, 0.90-0.95). Prognostic analysis showed that higher levels of circulating exosomal GPC-1 were associated with poorer OS in PC patients, and the combined HR for OS was 4.59 (random-effects model, 95% CI = 1.17-18.03, P = .022). The results of both studies were robust and neither had publication bias. CONCLUSION Circulating exosomal GPC-1 may be used as a diagnostic and prognostic biomarker for PC. However, this result needs to be validated by further research using a larger sample size.
Collapse
Affiliation(s)
- Zengyun Qiao
- Department of Hepatobiliary Surgery, Dalian Municipal Central Hospital, Dalian, China
- China Medical University, Shenyang, China
| | - Enbo Wang
- Department of Hepatobiliary Surgery, Dalian Municipal Central Hospital, Dalian, China
| | - Boyang Bao
- Department of Hepatobiliary Surgery, Dalian Municipal Central Hospital, Dalian, China
- Dalian Medical University, Dalian, China
| | | | | | - Dong Wang
- Department of Hepatobiliary Surgery, Dalian Municipal Central Hospital, Dalian, China
- China Medical University, Shenyang, China
| | - Liu Yuan
- Department of Hepatobiliary Surgery, Dalian Municipal Central Hospital, Dalian, China
- China Medical University, Shenyang, China
| |
Collapse
|
2
|
Bugazia D, Al-Najjar E, Esmail A, Abdelrahim S, Abboud K, Abdelrahim A, Umoru G, Rayyan HA, Abudayyeh A, Al Moustafa AE, Abdelrahim M. Pancreatic ductal adenocarcinoma: the latest on diagnosis, molecular profiling, and systemic treatments. Front Oncol 2024; 14:1386699. [PMID: 39011469 PMCID: PMC11247645 DOI: 10.3389/fonc.2024.1386699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/30/2024] [Indexed: 07/17/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is currently the fourth leading cause of death in the United States and is expected to be ranked second in the next 10 years due to poor prognosis and a rising incidence. Distant metastatic PDAC is associated with the worst prognosis among the different phases of PDAC. The diagnostic options for PDAC are convenient and available for staging, tumor response evaluation, and management of resectable or borderline resectable PDAC. However, imaging is crucial in PDAC diagnosis, monitoring, resectability appraisal, and response evaluation. The advancement of medical technologies is evolving, hence the use of imaging in PDAC treatment options has grown as well as the utilization of ctDNA as a tumor marker. Treatment options for metastatic PDAC are minimal with the primary goal of therapy limited to symptom relief or palliation, especially in patients with low functional capacity at the point of diagnosis. Molecular profiling has shown promising potential solutions that would push the treatment boundaries for patients with PDAC. In this review, we will discuss the latest updates from evidence-based guidelines regarding diagnosis, therapy response evaluation, prognosis, and surveillance, as well as illustrating novel therapies that have been recently investigated for PDAC, in addition to discussing the molecular profiling advances in PDAC.
Collapse
Affiliation(s)
- Doaa Bugazia
- Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Ebtesam Al-Najjar
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX, United States
| | - Abdullah Esmail
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX, United States
| | - Saifudeen Abdelrahim
- Challenge Early College HS, Houston Community College, Houston, TX, United States
| | - Karen Abboud
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX, United States
| | | | - Godsfavour Umoru
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX, United States
| | - Hashem A Rayyan
- Department of Medicine, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Ala Abudayyeh
- Section of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Maen Abdelrahim
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
3
|
Shi X. Significant elevation of serum CA19-9 and CA242 levels induced by dulaglutide. BMJ Case Rep 2024; 17:e257657. [PMID: 38719260 PMCID: PMC11085790 DOI: 10.1136/bcr-2023-257657] [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] [Indexed: 05/13/2024] Open
Abstract
The use of dulaglutide, a common medication for managing type 2 diabetes, rarely causes elevated pancreatic tumour markers. Here, we report the case of a woman in her mid-60s with diabetes for over 10 years. The patient presented with markedly elevated serum CA19-9 and CA242 levels revealed during a routine health examination despite being asymptomatic. She had been receiving dulaglutide injections for 16 months. Imaging and interventional assessments did not reveal any hepatobiliary, gastrointestinal or pancreatic neoplasm. After excluding alternate diagnoses, the patient was determined to exhibit an adverse reaction to dulaglutide use. Management involved the discontinuation of dulaglutide, which resulted in normalisation of serum CA19-9 and CA242 levels within 6 weeks. This case underscores the importance of discontinuing dulaglutide and monitoring changes in the biomarker levels in asymptomatic patients receiving dulaglutide, rather than immediately resorting to imaging and endoscopic examinations.
Collapse
Affiliation(s)
- Xiaomin Shi
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, China
| |
Collapse
|
4
|
Zhao F, Chen A, Wu X, Deng X, Yang J, Xue J. Heterogeneous changes in gut and tumor microbiota in patients with pancreatic cancer: insights from clinical evidence. BMC Cancer 2024; 24:478. [PMID: 38622651 PMCID: PMC11020926 DOI: 10.1186/s12885-024-12202-z] [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] [Received: 10/31/2023] [Accepted: 03/28/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Pancreatic cancer is the foremost contributor to cancer-related deaths globally, and its prevalence continues to rise annually. Nevertheless, the underlying mechanisms behind its development remain unclear and necessitate comprehensive investigation. METHODS In this study, a total of 29 fresh stool samples were collected from patients diagnosed with pancreatic cancer. The gut microbial data of healthy controls were obtained from the SRA database (SRA data number: SRP150089). Additionally, 28 serum samples and diseased tissues were collected from 14 patients with confirmed pancreatic cancer and 14 patients with chronic pancreatitis. Informed consent was obtained from both groups of patients. Microbial sequencing was performed using 16s rRNA. RESULTS The results showed that compared with healthy controls, the species abundance index of intestinal flora in patients with pancreatic cancer was increased (P < 0.05), and the number of beneficial bacteria at the genus level was reduced (P < 0.05). Compared with patients with chronic pancreatitis, the expression levels of CA242 and CA199 in the serum of patients with pancreatic cancer were increased (P < 0.05). The bacterial richness index of tumor microorganisms in patients with pancreatic cancer increased, while the diversity index decreased(P < 0.05). Furthermore, there was a change in the species composition at the genus level. Additionally, the expression level of CA242 was found to be significantly positively correlated with the relative abundance of Acinetobacter(P < 0.05). CONCLUSION Over all, the expression levels of serum tumor markers CA242 and CA19-9 in patients with pancreatic cancer are increased, while the beneficial bacteria in the intestine and tumor microenvironment are reduced and pathogenic bacteria are increased. Acinetobacter is a specific bacterial genus highly expressed in pancreatic cancer tissue.
Collapse
Affiliation(s)
- Feng Zhao
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, No. 55, Daxuecheng Middle Road, ShaPingBa District, 400016, Chongqing, People's Republic of China
| | - Anli Chen
- Department of Clinical Laboratory, University-Town Hospital of Chongqing Medical University, 401331, Chongqing, People's Republic of China
| | - Xiaotian Wu
- Department of Clinical Laboratory, University-Town Hospital of Chongqing Medical University, 401331, Chongqing, People's Republic of China
| | - Xiangyu Deng
- Department of Clinical Laboratory, University-Town Hospital of Chongqing Medical University, 401331, Chongqing, People's Republic of China
| | - Jiali Yang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, 401147, Chongqing, People's Republic of China.
| | - Jianjiang Xue
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, No. 55, Daxuecheng Middle Road, ShaPingBa District, 400016, Chongqing, People's Republic of China.
- Department of Clinical Laboratory, University-Town Hospital of Chongqing Medical University, 401331, Chongqing, People's Republic of China.
| |
Collapse
|
5
|
Shi W, Wartmann T, Accuffi S, Al-Madhi S, Perrakis A, Kahlert C, Link A, Venerito M, Keitel-Anselmino V, Bruns C, Croner RS, Zhao Y, Kahlert UD. Integrating a microRNA signature as a liquid biopsy-based tool for the early diagnosis and prediction of potential therapeutic targets in pancreatic cancer. Br J Cancer 2024; 130:125-134. [PMID: 37950093 PMCID: PMC10781694 DOI: 10.1038/s41416-023-02488-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 10/19/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
INTRODUCTION Pancreatic cancer is a highly aggressive cancer, and early diagnosis significantly improves patient prognosis due to the early implementation of curative-intent surgery. Our study aimed to implement machine-learning algorithms to aid in early pancreatic cancer diagnosis based on minimally invasive liquid biopsies. MATERIALS AND METHODS The analysis data were derived from nine public pancreatic cancer miRNA datasets and two sequencing datasets from 26 pancreatic cancer patients treated in our medical center, featuring small RNAseq data for patient-matched tumor and non-tumor samples and serum. Upon batch-effect removal, systematic analyses for differences between paired tissue and serum samples were performed. The robust rank aggregation (RRA) algorithm was used to reveal feature markers that were co-expressed by both sample types. The repeatability and real-world significance of the enriched markers were then determined by validating their expression in our patients' serum. The top candidate markers were used to assess the accuracy of predicting pancreatic cancer through four machine learning methods. Notably, these markers were also applied for the identification of pancreatic cancer and pancreatitis. Finally, we explored the clinical prognostic value, candidate targets and predict possible regulatory cell biology mechanisms involved. RESULTS Our multicenter analysis identified hsa-miR-1246, hsa-miR-205-5p, and hsa-miR-191-5p as promising candidate serum biomarkers to identify pancreatic cancer. In the test dataset, the accuracy values of the prediction model applied via four methods were 94.4%, 84.9%, 82.3%, and 83.3%, respectively. In the real-world study, the accuracy values of this miRNA signatures were 82.3%, 83.5%, 79.0%, and 82.2. Moreover, elevated levels of these miRNAs were significant indicators of advanced disease stage and allowed the discrimination of pancreatitis from pancreatic cancer with an accuracy rate of 91.5%. Elevated expression of hsa-miR-205-5p, a previously undescribed blood marker for pancreatic cancer, is associated with negative clinical outcomes in patients. CONCLUSION A panel of three miRNAs was developed with satisfactory statistical and computational performance in real-world data. Circulating hsa-miRNA 205-5p serum levels serve as a minimally invasive, early detection tool for pancreatic cancer diagnosis and disease staging and might help monitor therapy success.
Collapse
Affiliation(s)
- Wenjie Shi
- Molecular and Experimental Surgery, Faculty of Medicine and University Hospital Magdeburg, Department of General-, Visceral-, Vascular- and Transplant- Surgery, University of Magdeburg, Magdeburg, Germany
| | - Thomas Wartmann
- Molecular and Experimental Surgery, Faculty of Medicine and University Hospital Magdeburg, Department of General-, Visceral-, Vascular- and Transplant- Surgery, University of Magdeburg, Magdeburg, Germany
| | - Sara Accuffi
- Molecular and Experimental Surgery, Faculty of Medicine and University Hospital Magdeburg, Department of General-, Visceral-, Vascular- and Transplant- Surgery, University of Magdeburg, Magdeburg, Germany
| | - Sara Al-Madhi
- Molecular and Experimental Surgery, Faculty of Medicine and University Hospital Magdeburg, Department of General-, Visceral-, Vascular- and Transplant- Surgery, University of Magdeburg, Magdeburg, Germany
| | - Aristotelis Perrakis
- Molecular and Experimental Surgery, Faculty of Medicine and University Hospital Magdeburg, Department of General-, Visceral-, Vascular- and Transplant- Surgery, University of Magdeburg, Magdeburg, Germany
| | - Christoph Kahlert
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital Magdeburg, 39120, Magdeburg, Germany
| | - Marino Venerito
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital Magdeburg, 39120, Magdeburg, Germany
| | - Verena Keitel-Anselmino
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital Magdeburg, 39120, Magdeburg, Germany
| | - Christiane Bruns
- Faculty of Medicine and University Hospital Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Roland S Croner
- Molecular and Experimental Surgery, Faculty of Medicine and University Hospital Magdeburg, Department of General-, Visceral-, Vascular- and Transplant- Surgery, University of Magdeburg, Magdeburg, Germany
| | - Yue Zhao
- Faculty of Medicine and University Hospital Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany.
| | - Ulf D Kahlert
- Molecular and Experimental Surgery, Faculty of Medicine and University Hospital Magdeburg, Department of General-, Visceral-, Vascular- and Transplant- Surgery, University of Magdeburg, Magdeburg, Germany.
| |
Collapse
|
6
|
Szymoński K, Skirlińska-Nosek K, Lipiec E, Sofińska K, Czaja M, Wilkosz N, Krupa M, Wanat F, Ulatowska-Białas M, Adamek D. Combined analytical approach empowers precise spectroscopic interpretation of subcellular components of pancreatic cancer cells. Anal Bioanal Chem 2023; 415:7281-7295. [PMID: 37906289 PMCID: PMC10684650 DOI: 10.1007/s00216-023-04997-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 11/02/2023]
Abstract
The lack of specific and sensitive early diagnostic options for pancreatic cancer (PC) results in patients being largely diagnosed with late-stage disease, thus inoperable and burdened with high mortality. Molecular spectroscopic methodologies, such as Raman or infrared spectroscopies, show promise in becoming a leader in screening for early-stage cancer diseases, including PC. However, should such technology be introduced, the identification of differentiating spectral features between various cancer types is required. This would not be possible without the precise extraction of spectra without the contamination by necrosis, inflammation, desmoplasia, or extracellular fluids such as mucous that surround tumor cells. Moreover, an efficient methodology for their interpretation has not been well defined. In this study, we compared different methods of spectral analysis to find the best for investigating the biomolecular composition of PC cells cytoplasm and nuclei separately. Sixteen PC tissue samples of main PC subtypes (ductal adenocarcinoma, intraductal papillary mucinous carcinoma, and ampulla of Vater carcinoma) were collected with Raman hyperspectral mapping, resulting in 191,355 Raman spectra and analyzed with comparative methodologies, specifically, hierarchical cluster analysis, non-negative matrix factorization, T-distributed stochastic neighbor embedding, principal components analysis (PCA), and convolutional neural networks (CNN). As a result, we propose an innovative approach to spectra classification by CNN, combined with PCA for molecular characterization. The CNN-based spectra classification achieved over 98% successful validation rate. Subsequent analyses of spectral features revealed differences among PC subtypes and between the cytoplasm and nuclei of their cells. Our study establishes an optimal methodology for cancer tissue spectral data classification and interpretation that allows precise and cognitive studies of cancer cells and their subcellular components, without mixing the results with cancer-surrounding tissue. As a proof of concept, we describe findings that add to the spectroscopic understanding of PC.
Collapse
Affiliation(s)
- Krzysztof Szymoński
- Department of Pathomorphology, Medical College, Jagiellonian University, Kraków, Poland.
- Department of Pathomorphology, University Hospital, Kraków, Poland.
| | - Katarzyna Skirlińska-Nosek
- Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Jagiellonian University, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Ewelina Lipiec
- Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Jagiellonian University, Kraków, Poland
| | - Kamila Sofińska
- Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Jagiellonian University, Kraków, Poland
| | - Michał Czaja
- Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Jagiellonian University, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Natalia Wilkosz
- Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Jagiellonian University, Kraków, Poland
- AGH University of Krakow, Faculty of Physics and Applied Computer Science, Kraków, Poland
| | - Matylda Krupa
- Department of Pathomorphology, Medical College, Jagiellonian University, Kraków, Poland
| | - Filip Wanat
- Department of Pathomorphology, Medical College, Jagiellonian University, Kraków, Poland
| | - Magdalena Ulatowska-Białas
- Department of Pathomorphology, Medical College, Jagiellonian University, Kraków, Poland
- Department of Pathomorphology, University Hospital, Kraków, Poland
| | - Dariusz Adamek
- Department of Pathomorphology, Medical College, Jagiellonian University, Kraków, Poland
| |
Collapse
|
7
|
Zhu J, Li H. Serum expression of tumor marker CA242 in patients with different gynecological diseases. Lab Med 2023; 54:613-617. [PMID: 37035887 DOI: 10.1093/labmed/lmad017] [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: 04/11/2023] Open
Abstract
OBJECTIVE The aim of this study was to investigate the serum levels of CA242 in different types of gynecological diseases and its clinical significance. METHODS A total of 1021 patients with gynecological diseases and 499 healthy female controls were included in the study. The serum CA242 levels were detected and median value, -log10P value, and positive rate were calculated. Serum CA125 and HE4 levels of patients with ovarian lesions were measured, and the predictive value for ovarian cancer was statistically analyzed. RESULTS Higher serum CA242 levels were observed in patients with mature teratoma, ovarian cancer, and other gynecological tumor diseases than in healthy controls. In contrast, the CA242 levels in patients with cervical intraepithelial neoplasia, uterine polyps, or endometrial hyperplasia were comparable to that of controls. Moreover, serum CA242 expression was increased in malignant uterine and ovarian diseases compared with benign ones (P < .05). Specifically, combining CA242, CA125, and HE4 yielded a higher area under the receiver operating characteristic curve than single biomarkers (P < .05). CONCLUSION Heterogeneous increases in tumor marker CA242 expression levels are observed in different gynecological diseases, suggesting its potential value for clinical diagnosis.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Laboratory Medicine, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huidan Li
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai, China
| |
Collapse
|
8
|
Limanówka P, Ochman B, Świętochowska E. PiRNA Obtained through Liquid Biopsy as a Possible Cancer Biomarker. Diagnostics (Basel) 2023; 13:diagnostics13111895. [PMID: 37296747 DOI: 10.3390/diagnostics13111895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/21/2023] [Accepted: 05/27/2023] [Indexed: 06/12/2023] Open
Abstract
In recent years PIWI-interacting RNAs (piRNAs) have gained the interest of scientists, mainly because of their possible implications in cancer. Many kinds of research showed how their expression can be linked to malignant diseases. However, most of them evaluated the expression of piRNAs in tumor tissues. It was shown how these non-coding RNAs can interfere with many signaling pathways involved in the regulation of proliferation or apoptosis. A comparison of piRNA expression in tumor tissue and adjacent healthy tissues has demonstrated they can be used as biomarkers. However, this way of obtaining samples has a significant drawback, which is the invasiveness of such a procedure. Liquid biopsy is an alternative for acquiring biological material with little to no harm to a patient. Several different piRNAs in various types of cancer were shown to be expressed in bodily fluids such as blood or urine. Furthermore, their expression significantly differed between cancer patients and healthy individuals. Hence, this review aimed to assess the possible use of liquid biopsy for cancer diagnosis with piRNAs as biomarkers.
Collapse
Affiliation(s)
- Piotr Limanówka
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-800 Zabrze, Poland
| | - Błażej Ochman
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-800 Zabrze, Poland
| | - Elżbieta Świętochowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-800 Zabrze, Poland
| |
Collapse
|
9
|
Li B, Li Y, Li C, Yang J, Liu D, Wang H, Xu R, Zhang Y, Wei Q. An ultrasensitive split-type electrochemical immunosensor based on controlled-release strategy for detection of CA19-9. Biosens Bioelectron 2023; 227:115180. [PMID: 36858021 DOI: 10.1016/j.bios.2023.115180] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 02/09/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023]
Abstract
In this study, a novel split-type electrochemical immunosensor based on controlled release strategy was proposed for sensitive analysis and detection of tumor marker carbohydrate antigen 199 (CA19-9). Specifically, glucose (Glu) was encapsulated in carrier mesoporous silica (MSN) with encapsulation technology, and surface functionalized Zinc sulfide (ZnS) caps were used as "gatekeepers". The complex is formed by encapsulating Glu within MSN with ZnS (ZnS@MSN-Glu) as a signal amplifier labeled on the signal antibody (Ab2). And the Ab2 can detect the presence of antibodies. To reduce the interference of biological analysis, the immune recognition process of ZnS@MSN-Glu-Ab2 bioconjugate and antigen was carried out in 96-well microplate, which did not interfere with the electrochemical analysis process. Therefore, the low sensitivity detection caused by biofouling of nanomaterials and immunoreaction on the testing platform is eliminated. Subsequently, the opening and timed release of mesopores were controlled by external stimuli, the disulfide bond cleavage by dithiothreitol (DTT), and glucose was effectively released. Then nickel cobalt layered double hydroxide (NiCo-LDH) were directly hydrothermally grown on carbon cloth (CC) electrodeposited with copper selenide (CuSe) nanosheets to construct three-dimensional (3D) cactus-like NiCo-LDH/CuSe/CC sensing platform. It can realize the catalytic oxidation of released glucose, triggering glucose-mediated signal amplification. The synergistic effect of the 3D cactus structure and active nanomaterials promotes electron conduction. Taking the detection of carbohydrate antigen CA19-9 as an example, the immunosensor shows a wide linear concentration range (0.001-100 U/mL) with the limit of detection of 0.0005 U/mL, realizing highly sensitive detection of CA19-9. This biosensing technique has considerable advantages and provides an innovative approach for trace detection of other biomarkers.
Collapse
Affiliation(s)
- Bing Li
- Provincial Key Laboratory of Rural Energy Engineering in Yunnan, Yunnan Normal University, Kunming, 650500, PR China; Collaborative Innovation Center for Green Chemical Manufacturing and Accurate Detection, Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, PR China
| | - Yunxiao Li
- Provincial Key Laboratory of Rural Energy Engineering in Yunnan, Yunnan Normal University, Kunming, 650500, PR China
| | - Chenchen Li
- Collaborative Innovation Center for Green Chemical Manufacturing and Accurate Detection, Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, PR China
| | - Jinghui Yang
- Provincial Key Laboratory of Rural Energy Engineering in Yunnan, Yunnan Normal University, Kunming, 650500, PR China
| | - Deling Liu
- Collaborative Innovation Center for Green Chemical Manufacturing and Accurate Detection, Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, PR China
| | - Huabin Wang
- Provincial Key Laboratory of Rural Energy Engineering in Yunnan, Yunnan Normal University, Kunming, 650500, PR China
| | - Rui Xu
- Provincial Key Laboratory of Rural Energy Engineering in Yunnan, Yunnan Normal University, Kunming, 650500, PR China
| | - Yong Zhang
- Provincial Key Laboratory of Rural Energy Engineering in Yunnan, Yunnan Normal University, Kunming, 650500, PR China; Collaborative Innovation Center for Green Chemical Manufacturing and Accurate Detection, Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, PR China.
| | - Qin Wei
- Collaborative Innovation Center for Green Chemical Manufacturing and Accurate Detection, Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, PR China
| |
Collapse
|
10
|
Szymoński K, Chmura Ł, Lipiec E, Adamek D. Vibrational spectroscopy – are we close to finding a solution for early pancreatic cancer diagnosis? World J Gastroenterol 2023; 29:96-109. [PMID: 36683712 PMCID: PMC9850953 DOI: 10.3748/wjg.v29.i1.96] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/03/2022] [Accepted: 10/31/2022] [Indexed: 01/04/2023] Open
Abstract
Pancreatic cancer (PC) is an aggressive and lethal neoplasm, ranking seventh in the world for cancer deaths, with an overall 5-year survival rate of below 10%. The knowledge about PC pathogenesis is rapidly expanding. New aspects of tumor biology, including its molecular and morphological heterogeneity, have been reported to explain the complicated “cross-talk” that occurs between the cancer cells and the tumor stroma or the nature of pancreatic ductal adenocarcinoma-associated neural remodeling. Nevertheless, currently, there are no specific and sensitive diagnosis options for PC. Vibrational spectroscopy (VS) shows a promising role in the development of early diagnosis technology. In this review, we summarize recent reports about improvements in spectroscopic methodologies, briefly explain and highlight the drawbacks of each of them, and discuss available solutions. The important aspects of spectroscopic data evaluation with multivariate analysis and a convolutional neural network methodology are depicted. We conclude by presenting a study design for systemic verification of the VS-based methods in the diagnosis of PC.
Collapse
Affiliation(s)
- Krzysztof Szymoński
- Department of Pathomorphology, Jagiellonian University Medical College, Cracow 33-332, Poland
- Department of Pathomorphology, University Hospital in Cracow, Cracow 31-501, Poland
| | - Łukasz Chmura
- Department of Pathomorphology, Jagiellonian University Medical College, Cracow 33-332, Poland
- Department of Pathomorphology, University Hospital in Cracow, Cracow 31-501, Poland
| | - Ewelina Lipiec
- M. Smoluchowski Institute of Physics, Jagiellonian University, Cracow 30-348, Poland
| | - Dariusz Adamek
- Department of Pathomorphology, University Hospital in Cracow, Cracow 31-501, Poland
- Department of Neuropathology, Jagiellonian University Medical College, Cracow 33-332, Poland
| |
Collapse
|
11
|
Identification of the Genetic Association Between Type-2-Diabetes and Pancreatic Cancer. Biochem Genet 2022; 61:1143-1162. [DOI: 10.1007/s10528-022-10308-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022]
|
12
|
Perelló-Reus CM, Rubio-Tomás T, Cisneros-Barroso E, Ibargüen-González L, Segura-Sampedro JJ, Morales-Soriano R, Barceló C. Challenges in precision medicine in pancreatic cancer: A focus in cancer stem cells and microbiota. Front Oncol 2022; 12:995357. [PMID: 36531066 PMCID: PMC9751445 DOI: 10.3389/fonc.2022.995357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/15/2022] [Indexed: 12/03/2022] Open
Abstract
Pancreatic cancer adenocarcinoma (PDAC) is a lethal disease, with the lowest 5-years survival rate of all cancers due to late diagnosis. Despite the advance and success of precision oncology in gastrointestinal cancers, the frequency of molecular-informed therapy decisions in PDAC is currently neglectable. The reasons for this dismal situation are mainly the absence of effective early diagnostic biomarkers and therapy resistance. PDAC cancer stem cells (PDAC-SC), which are regarded as essential for tumor initiation, relapse and drug resistance, are highly dependent on their niche i.e. microanatomical structures of the tumor microenvironment. There is an altered microbiome in PDAC patients embedded within the highly desmoplastic tumor microenvironment, which is known to determine therapeutic responses and affecting survival in PDAC patients. We consider that understanding the communication network that exists between the microbiome and the PDAC-SC niche by co-culture of patient-derived organoids (PDOs) with TME microbiota would recapitulate the complexity of PDAC paving the way towards a precision oncology treatment-response prediction.
Collapse
Affiliation(s)
- Catalina M. Perelló-Reus
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases (HUSE), Palma de Mallorca, Spain,*Correspondence: Carles Barceló, ; Catalina M. Perelló-Reus,
| | | | | | - Lesly Ibargüen-González
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases (HUSE), Palma de Mallorca, Spain
| | - Juan José Segura-Sampedro
- Advanced Oncological Surgery, Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain,General and Digestive Surgery Unit, Hospital Universitari Son Espases, School of Medicine, Balearic Islands Health Research Institute, University of Balearic Islands, Palma de Mallorca, Spain
| | - Rafael Morales-Soriano
- Advanced Oncological Surgery, Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain,General and Digestive Surgery Unit, Hospital Universitari Son Espases, School of Medicine, Balearic Islands Health Research Institute, University of Balearic Islands, Palma de Mallorca, Spain
| | - Carles Barceló
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases (HUSE), Palma de Mallorca, Spain,*Correspondence: Carles Barceló, ; Catalina M. Perelló-Reus,
| |
Collapse
|
13
|
Nakamura K, Zhu Z, Roy S, Jun E, Han H, Munoz RM, Nishiwada S, Sharma G, Cridebring D, Zenhausern F, Kim S, Roe DJ, Darabi S, Han IW, Evans DB, Yamada S, Demeure MJ, Becerra C, Celinski SA, Borazanci E, Tsai S, Kodera Y, Park JO, Bolton JS, Wang X, Kim SC, Von Hoff D, Goel A. An Exosome-based Transcriptomic Signature for Noninvasive, Early Detection of Patients With Pancreatic Ductal Adenocarcinoma: A Multicenter Cohort Study. Gastroenterology 2022; 163:1252-1266.e2. [PMID: 35850192 PMCID: PMC9613527 DOI: 10.1053/j.gastro.2022.06.090] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/18/2022] [Accepted: 06/25/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinoma (PDAC) incidence is rising worldwide, and most patients present with an unresectable disease at initial diagnosis. Measurement of carbohydrate antigen 19-9 (CA19-9) levels lacks adequate sensitivity and specificity for early detection; hence, there is an unmet need to develop alternate molecular diagnostic biomarkers for PDAC. Emerging evidence suggests that tumor-derived exosomal cargo, particularly micro RNAs (miRNAs), offer an attractive platform for the development of cancer-specific biomarkers. Herein, genomewide profiling in blood specimens was performed to develop an exosome-based transcriptomic signature for noninvasive and early detection of PDAC. METHODS Small RNA sequencing was undertaken in a cohort of 44 patients with an early-stage PDAC and 57 nondisease controls. Using machine-learning algorithms, a panel of cell-free (cf) and exosomal (exo) miRNAs were prioritized that discriminated patients with PDAC from control subjects. Subsequently, the performance of the biomarkers was trained and validated in independent cohorts (n = 191) using quantitative reverse transcription polymerase chain reaction (qRT-PCR) assays. RESULTS The sequencing analysis initially identified a panel of 30 overexpressed miRNAs in PDAC. Subsequently using qRT-PCR assays, the panel was reduced to 13 markers (5 cf- and 8 exo-miRNAs), which successfully identified patients with all stages of PDAC (area under the curve [AUC] = 0.98 training cohort; AUC = 0.93 validation cohort); but more importantly, was equally robust for the identification of early-stage PDAC (stages I and II; AUC = 0.93). Furthermore, this transcriptomic signature successfully identified CA19-9 negative cases (<37 U/mL; AUC = 0.96), when analyzed in combination with CA19-9 levels, significantly improved the overall diagnostic accuracy (AUC = 0.99 vs AUC = 0.86 for CA19-9 alone). CONCLUSIONS In this study, an exosome-based liquid biopsy signature for the noninvasive and robust detection of patients with PDAC was developed.
Collapse
Affiliation(s)
- Kota Nakamura
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Monrovia, California
| | - Zhongxu Zhu
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Monrovia, California; Department of Surgery, The Chinese University of Hong Kong. Prince of Wales Hospital, Shatin, N.T., Hong Kong, SAR, China; Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Souvick Roy
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Monrovia, California
| | - Eunsung Jun
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Ulsan University College of Medicine and Asan Medical Center, Seoul, Korea
| | - Haiyong Han
- The Translational Genomics Research Institute, Phoenix, Arizona
| | - Ruben M Munoz
- The Translational Genomics Research Institute, Phoenix, Arizona
| | - Satoshi Nishiwada
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Monrovia, California
| | - Geeta Sharma
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Monrovia, California
| | | | - Frederic Zenhausern
- Center for Applied NanoBioscience and Medicine, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona
| | - Seungchan Kim
- Department of Electrical and Computer Engineering, Roy G. Perry College of Engineering, Prairie View A&M University, Prairie View, Texas
| | - Denise J Roe
- Department of Epidemiology and Biostatistics, The University of Arizona, Tucson, Arizona
| | - Sourat Darabi
- Hoag Family Center Institute, Newport Beach, California
| | - In-Woong Han
- Division of Hepato-Biliary Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Douglas B Evans
- Department of Surgery, The Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Suguru Yamada
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Michael J Demeure
- The Translational Genomics Research Institute, Phoenix, Arizona; Hoag Family Center Institute, Newport Beach, California
| | - Carlos Becerra
- Baylor Scott and White Research Institute, Baylor University Medical Center, Dallas, Texas
| | - Scott A Celinski
- Baylor Scott and White Research Institute, Baylor University Medical Center, Dallas, Texas
| | | | - Susan Tsai
- Department of Surgery, The Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Joon Oh Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - John S Bolton
- Department of Surgery, Ochsner Clinic Foundation, New Orleans, Louisiana
| | - Xin Wang
- Department of Surgery, The Chinese University of Hong Kong. Prince of Wales Hospital, Shatin, N.T., Hong Kong, SAR, China.
| | - Song Cheol Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Ulsan University College of Medicine and Asan Medical Center, Seoul, Korea.
| | - Daniel Von Hoff
- The Translational Genomics Research Institute, Phoenix, Arizona.
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Monrovia, California; City of Hope Comprehensive Cancer Center, Duarte, California.
| |
Collapse
|
14
|
Su J, Wang Y, Shao H, You X, Li S. Value of multi-detector computed tomography combined with serum tumor markers in diagnosis, preoperative, and prognostic evaluation of pancreatic cancer. World J Surg Oncol 2022; 20:323. [PMID: 36175918 PMCID: PMC9520929 DOI: 10.1186/s12957-022-02785-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/18/2022] [Indexed: 12/02/2022] Open
Abstract
Background Multi-detector computed tomography (MDCT) and serum tumor markers are commonly used in the diagnosis of pancreatic cancer (PC). In this article, we focused on the evaluation of the clinical value of MDCT combined with serum tumor markers CA199, CA242, and CEA in diagnosis, preoperative, and prognostic evaluation of PC. Methods Eighty-five PC patients (PC group) and 39 patients with pancreatitis (control group) admitted to our hospital were selected for our present research study. MDCT, CA199, CA242, and CEA examination were examined in all patients, and their value in diagnosis, preoperative, and prognostic evaluation of PC was retrospectively analyzed. Results There were 69 patients whose clinical staging results of MDCT were consistent with the postoperative pathological diagnosis. The coincidence rate was 70.00% in stage I, 62.96% in stage II, 72.72% in stage III, and 80.00% in stage IV, respectively, and the overall coincidence rate was 69.57%The levels of CA199, CA242, and CEA in PC group were remarkably higher than those in control group and were sharply correlated with clinical stage, differentiation degree, and distant metastasis. The sensitivity, accuracy, and negative predictive value of MDCT combined with serum CA199, CA242 and CEA in the diagnosis of PC were significantly improved compared with those of each single test. In PC group, the 2-year event-free survival rate of the group with high CA199, CA242, and CEA expression was remarkably lower than that of the low expression group. Conclusion MDCT combined with CA199, CA242, and CEA notably improved the diagnostic efficiency of PC and had guiding significance for preoperative and prognostic evaluation of PC.
Collapse
Affiliation(s)
- Jianli Su
- Department of Clinical Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - Yunfeng Wang
- Department of Clinical Laboratory, Chengyang People's Hospital, Qingdao, 266109, China
| | - Hua Shao
- Radiophysics Department, The Affiliated Qingdao Central Hospital of Qingdao University, The Second Affiliated Hospital of Medical College of Qingdao University, Qingdao, 266042, China
| | - Xinting You
- Department of Endoscopic Diagnosis and Treatment, Qingdao Eighth People's Hospital, Qingdao, 266100, China
| | - Shuying Li
- Department of Hepatobiliary Pancreatic Surgery (I), Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China.
| |
Collapse
|
15
|
Balaban DV, Marin FS, Manucu G, Zoican A, Ciochina M, Mina V, Patoni C, Vladut C, Bucurica S, Costache RS, Ionita-Radu F, Jinga M. Clinical characteristics and outcomes in carbohydrate antigen 19-9 negative pancreatic cancer. World J Clin Oncol 2022; 13:630-640. [PMID: 36157158 PMCID: PMC9346420 DOI: 10.5306/wjco.v13.i7.630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/19/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of death from cancer worldwide. Tumor markers like carbohydrate antigen 19-9 (CA 19-9) have been proven valuable as a diagnostic tool and a predictor for tumor staging and response to therapy.
AIM To delineate the phenotype of normal CA 19-9 PDAC according to clinical features, disease staging and prognosis as compared with high CA 19-9 PDAC cases.
METHODS We performed a retrospective single-center analysis of all PDAC cases admitted in our Gastroenterology department over a period of 30 mo that were diagnosed by endoscopic ultrasound-guided tissue acquisition. Patients were divided into two groups according to CA 19-9 levels over a threshold of 37 U/mL. We performed a comparison between the two groups with regard to demographic and clinical data, biomarkers, tumor staging and 6-mo survival.
RESULTS Altogether 111 patients were recruited with 29 having documented normal CA 19-9 (< 37 U/mL). In the CA 19-9 negative group of patients, 20.68% had elevated levels of both CEA and CA 125, 13.79% for CA 125 only whilst 17.24% for CEA only. The two groups had similar demographic characteristics. Abdominal pain was more frequently reported in positive vs negative CA 19-9 PDAC cases (76.83% vs 55.17%), while smoking was slightly more prevalent in the latter group (28.04% vs 31.03%). Tumors over 2 cm were more frequently seen in the positive CA 19-9 group, reflecting a higher proportion of locally advanced and metastatic neoplasia (87.7% vs 79.3%). Six-month survival was higher for the negative CA 19-9 group (58.62% vs 47.56%).
CONCLUSION Elevated CA 19-9 at diagnosis seems to be associated with a more pronounced symptomatology, high tumor burden and poor prognosis compared to negative CA 19-9 PDAC cases. CEA and CA 125 can be adjunctive useful markers for PDAC, especially in CA 19-9 negative cases.
Collapse
Affiliation(s)
- Daniel Vasile Balaban
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Flavius Stefan Marin
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
- Department of Gastroenterology and Digestive Oncology, Hôpital Cochin, Paris 75014, France
| | - George Manucu
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Andreea Zoican
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Marina Ciochina
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Victor Mina
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Cristina Patoni
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Catalina Vladut
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Prof Dr. Agrippa Ionescu Clinical Emergency Hospital, Bucharest 020021, Romania
| | - Sandica Bucurica
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Raluca Simona Costache
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Florentina Ionita-Radu
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Mariana Jinga
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| |
Collapse
|
16
|
Kartal E, Schmidt TSB, Molina-Montes E, Rodríguez-Perales S, Wirbel J, Maistrenko OM, Akanni WA, Alashkar Alhamwe B, Alves RJ, Carrato A, Erasmus HP, Estudillo L, Finkelmeier F, Fullam A, Glazek AM, Gómez-Rubio P, Hercog R, Jung F, Kandels S, Kersting S, Langheinrich M, Márquez M, Molero X, Orakov A, Van Rossum T, Torres-Ruiz R, Telzerow A, Zych K, Benes V, Zeller G, Trebicka J, Real FX, Malats N, Bork P. A faecal microbiota signature with high specificity for pancreatic cancer. Gut 2022; 71:1359-1372. [PMID: 35260444 PMCID: PMC9185815 DOI: 10.1136/gutjnl-2021-324755] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 12/05/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Recent evidence suggests a role for the microbiome in pancreatic ductal adenocarcinoma (PDAC) aetiology and progression. OBJECTIVE To explore the faecal and salivary microbiota as potential diagnostic biomarkers. METHODS We applied shotgun metagenomic and 16S rRNA amplicon sequencing to samples from a Spanish case-control study (n=136), including 57 cases, 50 controls, and 29 patients with chronic pancreatitis in the discovery phase, and from a German case-control study (n=76), in the validation phase. RESULTS Faecal metagenomic classifiers performed much better than saliva-based classifiers and identified patients with PDAC with an accuracy of up to 0.84 area under the receiver operating characteristic curve (AUROC) based on a set of 27 microbial species, with consistent accuracy across early and late disease stages. Performance further improved to up to 0.94 AUROC when we combined our microbiome-based predictions with serum levels of carbohydrate antigen (CA) 19-9, the only current non-invasive, Food and Drug Administration approved, low specificity PDAC diagnostic biomarker. Furthermore, a microbiota-based classification model confined to PDAC-enriched species was highly disease-specific when validated against 25 publicly available metagenomic study populations for various health conditions (n=5792). Both microbiome-based models had a high prediction accuracy on a German validation population (n=76). Several faecal PDAC marker species were detectable in pancreatic tumour and non-tumour tissue using 16S rRNA sequencing and fluorescence in situ hybridisation. CONCLUSION Taken together, our results indicate that non-invasive, robust and specific faecal microbiota-based screening for the early detection of PDAC is feasible.
Collapse
Affiliation(s)
- Ece Kartal
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Collaboration for joint PhD degree, European Molecular Biology Laboratory and Heidelberg University, Heidelberg, Germany
| | - Thomas S B Schmidt
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Esther Molina-Montes
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC), Madrid, Spain
| | - Sandra Rodríguez-Perales
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC), Madrid, Spain
- Molecular Cytogenetics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Jakob Wirbel
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Collaboration for joint PhD degree, European Molecular Biology Laboratory and Heidelberg University, Heidelberg, Germany
| | - Oleksandr M Maistrenko
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Wasiu A Akanni
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Bilal Alashkar Alhamwe
- Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung School (UGMLC), Philipps University Marburg Faculty of Medicine, Marburg, Germany
| | - Renato J Alves
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Alfredo Carrato
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC), Madrid, Spain
- Medical Oncology Department of Oncology, Hospital Ramón y Cajal, Madrid, Spain
- University of Alcala de Henares, Alcala de Henares, Spain
| | - Hans-Peter Erasmus
- Translational Hepatology Department of Internal Medicine I, Goethe-Universitat Frankfurt am Main, Frankfurt am Main, Germany
| | - Lidia Estudillo
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC), Madrid, Spain
| | - Fabian Finkelmeier
- Translational Hepatology Department of Internal Medicine I, Goethe-Universitat Frankfurt am Main, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Hessen, Germany
| | - Anthony Fullam
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Anna M Glazek
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Paulina Gómez-Rubio
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC), Madrid, Spain
| | - Rajna Hercog
- Genomic Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Ferris Jung
- Genomic Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Stefanie Kandels
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Stephan Kersting
- Department of Surgery, Erlangen University Hospital, Erlangen, Germany
- Department of Surgery, University of Greifswald, Greifswald, Germany
| | | | - Mirari Márquez
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC), Madrid, Spain
| | - Xavier Molero
- Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Barcelona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Askarbek Orakov
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Thea Van Rossum
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Raul Torres-Ruiz
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC), Madrid, Spain
- Molecular Cytogenetics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Anja Telzerow
- Genomic Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Konrad Zych
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Vladimir Benes
- Genomic Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Georg Zeller
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jonel Trebicka
- Translational Hepatology Department of Internal Medicine I, Goethe-Universitat Frankfurt am Main, Frankfurt am Main, Germany
- EF Clif, European Foundation for the Study of Chronic Liver Failure, Barcelona, Spain
| | - Francisco X Real
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC), Madrid, Spain
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Nuria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC), Madrid, Spain
| | - Peer Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
- Yonsei Frontier Lab (YFL), Yonsei University, Seoul, South Korea
- Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| |
Collapse
|
17
|
Szymoński K, Milian-Ciesielska K, Lipiec E, Adamek D. Current Pathology Model of Pancreatic Cancer. Cancers (Basel) 2022; 14:2321. [PMID: 35565450 PMCID: PMC9105915 DOI: 10.3390/cancers14092321] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 02/01/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most aggressive and lethal malignant neoplasms, ranking in seventh place in the world in terms of the incidence of death, with overall 5-year survival rates still below 10%. The knowledge about PC pathomechanisms is rapidly expanding. Daily reports reveal new aspects of tumor biology, including its molecular and morphological heterogeneity, explain complicated "cross-talk" that happens between the cancer cells and tumor stroma, or the nature of the PC-associated neural remodeling (PANR). Staying up-to-date is hard and crucial at the same time. In this review, we are focusing on a comprehensive summary of PC aspects that are important in pathologic reporting, impact patients' outcomes, and bring meaningful information for clinicians. Finally, we show promising new trends in diagnostic technologies that might bring a difference in PC early diagnosis.
Collapse
Affiliation(s)
- Krzysztof Szymoński
- Department of Pathomorphology, Jagiellonian University Medical College, 31-531 Cracow, Poland;
- Department of Pathomorphology, University Hospital, 30-688 Cracow, Poland;
| | | | - Ewelina Lipiec
- M. Smoluchowski Institute of Physics, Jagiellonian University, 30-348 Cracow, Poland;
| | - Dariusz Adamek
- Department of Pathomorphology, Jagiellonian University Medical College, 31-531 Cracow, Poland;
| |
Collapse
|
18
|
Yang Y, Su X, Shen K, Zhang C, Dai H, Ma H, Jiang Y, Shuai L, Liu Z, You J, Min K, Chen Z. PUM1 is upregulated by DNA methylation to suppress antitumor immunity and results in poor prognosis in pancreatic cancer. Transl Cancer Res 2022; 10:2153-2168. [PMID: 35116535 PMCID: PMC8798770 DOI: 10.21037/tcr-20-3295] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/26/2021] [Indexed: 12/24/2022]
Abstract
Background Pancreatic carcinoma (PAAD) is a highly malignant cancer with a poor prognosis and high mortality rate. Pumilio homologous protein 1 (PUM1) promotes cell growth, invasion, and metastasis and suppresses apoptosis in many different kinds of cancers, such as non-small-cell lung carcinoma (NSCLC), ovarian cancer and lymphocyte leukemia. However, the underlying mechanism and potential role of PUM1 in PAAD have not been investigated. Methods Bioinformatics analysis was performed using multiple databases [The Cancer Genome Atlas (TCGA), Gene Expression Profiling Interactive Analysis (GEPIA), BBCancer, Human Protein Atlas (HPA), MethSurv, cBioPortal, The Cancer Imaging Archive (TCIA), xCell, Gene Expression Omnibus (GEO)] to explore the diagnostic and prognostic role of PUM1, and the relationship between expression of PUM1 and prognosis of patients with PAAD. The analysis was further validated using the Kaplan-Meier plotter. Results PUM1 plays a role in both diagnostic and prognostic prediction. The PUM1 mRNA expression level correlates with both the prognosis and incidence of pancreatic cancer. PUM1 can serve as a potential diagnostic indicator for pancreatic cancer. Furthermore, the DNA methylation levels of PUM1 affects its oncogene function in pancreatic cancer. PUM1 can also inhibit the immune microenvironment by altering immune cell infiltration, which affects immunotherapy response in pancreatic cancer. Conclusions PUM1 takes a crucial part in the immune microenvironment and immunotherapy response of PAAD and is potentially useful for the development of novel diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Yishi Yang
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Xingxing Su
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Kaicheng Shen
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Chengcheng Zhang
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Haisu Dai
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Hongbo Ma
- Department of Oncology, The Fuling Central Hospital of Chongqing City, Chongqing, China
| | - Yan Jiang
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Ling Shuai
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Zhipeng Liu
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Jinshan You
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Ke Min
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Zhiyu Chen
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| |
Collapse
|
19
|
Dharan M, Ryan-Fisher C. Sister Mary Joseph Nodule and Concomitant Pancreatitis as Initial Presentation of Pancreatic Adenocarcinoma - Case Report and Review of the Literature. Cureus 2021; 13:e20069. [PMID: 35003942 PMCID: PMC8723718 DOI: 10.7759/cureus.20069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2021] [Indexed: 11/28/2022] Open
Abstract
Umbilical metastasis [Sister Mary Joseph Nodule (SMJN)] is a rare presentation of visceral abdominal/pelvic malignancy. It is less commonly seen in metastatic pancreatic adenocarcinoma and there are only over a hundred cases to date in the literature on this topic. This article highlights a case of metastatic pancreatic adenocarcinoma presenting as SMJN and concomitant pancreatitis (which to the best of our knowledge is the first such report to date) with discussions regarding the etiopathogenesis of this phenomenon and presents a brief literature review on pancreatic adenocarcinoma presenting as SMJN.
Collapse
Affiliation(s)
- Murali Dharan
- Gastroenterology and Hepatology, University of Connecticut Health Center, Farmington, USA
| | | |
Collapse
|
20
|
Perales S, Torres C, Jimenez-Luna C, Prados J, Martinez-Galan J, Sanchez-Manas JM, Caba O. Liquid biopsy approach to pancreatic cancer. World J Gastrointest Oncol 2021; 13:1263-1287. [PMID: 34721766 PMCID: PMC8529923 DOI: 10.4251/wjgo.v13.i10.1263] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/18/2021] [Accepted: 08/27/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) continues to pose a major clinical challenge. There has been little improvement in patient survival over the past few decades, and it is projected to become the second leading cause of cancer mortality by 2030. The dismal 5-year survival rate of less than 10% after the diagnosis is attributable to the lack of early symptoms, the absence of specific biomarkers for an early diagnosis, and the inadequacy of available chemotherapies. Most patients are diagnosed when the disease has already metastasized and cannot be treated. Cancer interception is vital, actively intervening in the malignization process before the development of a full-blown advanced tumor. An early diagnosis of PC has a dramatic impact on the survival of patients, and improved techniques are urgently needed to detect and evaluate this disease at an early stage. It is difficult to obtain tissue biopsies from the pancreas due to its anatomical position; however, liquid biopsies are readily available and can provide useful information for the diagnosis, prognosis, stratification, and follow-up of patients with PC and for the design of individually tailored treatments. The aim of this review was to provide an update of the latest advances in knowledge on the application of carbohydrates, proteins, cell-free nucleic acids, circulating tumor cells, metabolome compounds, exosomes, and platelets in blood as potential biomarkers for PC, focusing on their clinical relevance and potential for improving patient outcomes.
Collapse
Affiliation(s)
- Sonia Perales
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Granada 18071, Spain
| | - Carolina Torres
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Sciences, University of Granada, Granada 18071, Spain
| | - Cristina Jimenez-Luna
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain
| | - Joaquina Martinez-Galan
- Department of Medical Oncology, Hospital Universitario Virgen de las Nieves, Granada 18011, Spain
| | | | - Octavio Caba
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain
| |
Collapse
|
21
|
O'Neill RS, Stoita A. Biomarkers in the diagnosis of pancreatic cancer: Are we closer to finding the golden ticket? World J Gastroenterol 2021; 27:4045-4087. [PMID: 34326612 PMCID: PMC8311531 DOI: 10.3748/wjg.v27.i26.4045] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/24/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is a leading cause of cancer related mortality on a global scale. The disease itself is associated with a dismal prognosis, partly due to its silent nature resulting in patients presenting with advanced disease at the time of diagnosis. To combat this, there has been an explosion in the last decade of potential candidate biomarkers in the research setting in the hope that a diagnostic biomarker may provide a glimmer of hope in what is otherwise quite a substantial clinical dilemma. Currently, serum carbohydrate antigen 19-9 is utilized in the diagnostic work-up of patients diagnosed with PC however this biomarker lacks the sensitivity and specificity associated with a gold-standard marker. In the search for a biomarker that is both sensitive and specific for the diagnosis of PC, there has been a paradigm shift towards a focus on liquid biopsy and the use of diagnostic panels which has subsequently proved to have efficacy in the diagnosis of PC. Currently, promising developments in the field of early detection on PC using diagnostic biomarkers include the detection of microRNA (miRNA) in serum and circulating tumour cells. Both these modalities, although in their infancy and yet to be widely accepted into routine clinical practice, possess merit in the early detection of PC. We reviewed over 300 biomarkers with the aim to provide an in-depth summary of the current state-of-play regarding diagnostic biomarkers in PC (serum, urinary, salivary, faecal, pancreatic juice and biliary fluid).
Collapse
Affiliation(s)
- Robert S O'Neill
- Department of Gastroenterology, St Vincent's Hospital Sydney, Sydney 2010, Australia
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney 2010, Australia
| | - Alina Stoita
- Department of Gastroenterology, St Vincent's Hospital Sydney, Sydney 2010, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney 2010, Australia
| |
Collapse
|
22
|
Abstract
Electrochemical immunosensors are affinity-based biosensors characterized by several useful features such as specificity, miniaturizability, low cost and simplicity, making them very interesting for many applications in several scientific fields. One of the significant issues in the design of electrochemical immunosensors is to increase the system’s sensitivity. Different strategies have been developed, one of the most common is the use of nanostructured materials as electrode materials, nanocarriers, electroactive or electrocatalytic nanotracers because of their abilities in signal amplification and biocompatibility. In this review, we will consider some of the most used nanostructures employed in the development of electrochemical immunosensors (e.g., metallic nanoparticles, graphene, carbon nanotubes) and many other still uncommon nanomaterials. Furthermore, their diagnostic applications in the last decade will be discussed, referring to two relevant issues of present-day: the detection of tumor markers and viruses.
Collapse
|
23
|
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]
|
24
|
Fan GC, Gu S, Zhang D, Hu Z, Luo X. Platinum-based nanocomposite as oxygen reduction catalyst for efficient signal amplification: Toward building of high-performance photocathodic immunoassay. Biosens Bioelectron 2020; 168:112563. [PMID: 32892117 DOI: 10.1016/j.bios.2020.112563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/13/2020] [Accepted: 08/24/2020] [Indexed: 10/23/2022]
Abstract
Photocathodic bioassays have shown great potential to apply in real bio-sample detection owning to their intrinsic abilities against interference from reductive species. However, the pursuit of photocathodic bioassays with excellent detection performance is still in the infancy. Herein, an advanced signal amplifier of platinum-based nanocatalyst with efficient oxygen reduction capability was explored to build a high-performance photocathodic immunoassay. The target model of carbohydrate antigen 19-9 (CA19-9, Ag) was used for describing the sensing platform. Specifically, the nontoxic Au/CuBi2O4 photocathode was first prepared by decorating Au nanoparticles on CuBi2O4 nanofilm and was used as the matrix to anchor capture CA19-9 antibody (Ab1). Platinum (Pt) nanoparticles were loaded on graphene (GR) nanosheet to form Pt/GR nanocomposite, which was utilized as signal amplifier conjugating with signal CA19-9 antibody (Ab2). When specific sandwich immunoreaction happened, the Pt/GR played the role of an efficient nanocatalyst to accelerate the reduction reaction of electron acceptor of oxygen in the electrolyte, causing evidently enhanced cathodic photocurrent signal. By incorporating this superior signal amplification strategy into the anti-interference photocathodic immunoassay, highly sensitive and specific detection of target Ag was realized. This work pioneers a new perspective for the design of advanced photocathodic bioanalysis for various targets of interest.
Collapse
Affiliation(s)
- Gao-Chao Fan
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Shiting Gu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Di Zhang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Ze Hu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Xiliang Luo
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China.
| |
Collapse
|
25
|
TNM staging for GIT cancers is correlated with the level of MMPs and TGF-β1. Clin Exp Med 2020; 20:545-555. [PMID: 32772210 DOI: 10.1007/s10238-020-00651-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 07/31/2020] [Indexed: 02/08/2023]
Abstract
Gastrointestinal (GIT) cancers represent the third common cancers worldwide, characterized by rapid progression and higher mortality rate. Matrix metalloproteinases (MMPs) play an important role in cancer metastases. The present study was conducted to estimate and evaluate the role of MMP-7, -9, -10 and -12 and TGF β1 along with conventional biomarkers (CEA and CA19-9) in gastric (GC), pancreatic (PC) and colorectal cancer (CRC) staging system according to tumor size (T), included lymph node (N) and metastasis (M). Seventy-five patients were divided into GC group (n = 25), PC group (n = 25), CRC group (n = 25) and twenty-five healthy subjects (control group). Serum levels of MMP-7, -10 and -12 were assayed simultaneously using luminex multiplex technique. Also, MMP-9, TGF-β1, CA19-9 and CEA were determined by ELISA. MMP-7,-9,-10, -12, TGF-β1 and CEA levels were significantly (p < 0.001) higher in GIT cancer groups compared with control. CA19-9 was significantly (p < 0.001) higher in PC and CRC groups compared with control. MMP-9 was positively correlated with TNM staging in PC patients. MMP-12 was negatively correlated with T in PC and positively correlated with M in CRC group. CA 19-9 was positively correlated with M grade in CRC. Depending on the estimated cutoff values of area under receiver curve; CA19-9 and MMP-7 were excellent diagnostic markers in PC, CEA and MMP-7 were excellent in CRC, and MMP-7 and MMP-9 were excellent in GC. Our findings indicated the clinical utility of MMPs in diagnosis and TNM staging of GIT cancers along with CEA and CA19-9.
Collapse
|
26
|
Li X, Li S, Liu L, Hong J, Zhao T, Gao C. Effect of Perioperative CEA and CA24-2 on Prognosis of Early Resectable Pancreatic Ductal Adenocarcinoma. J Cancer 2020; 11:9-15. [PMID: 31892968 PMCID: PMC6930410 DOI: 10.7150/jca.33767] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 09/16/2019] [Indexed: 12/22/2022] Open
Abstract
Patients with resectable pancreatic ductal adenocarcinoma (PDAC) show differential prognosis after radical resection. Currently, cancer grading and surgical criteria depend heavily on imaging and anatomical diagnosis. It's essential to set up a model with reliable prognostic factors during the perioperative period to assess prognosis in PDAC patients. In this study, 103 patients diagnosed with PDAC who underwent radical resection were recruited. The predictive value of preoperative carcinoembryonic antigen (CEA), postoperative CA24-2 and the combination of two for overall survival (OS) were evaluated. Both pre-CEA and post-CA24-2 were found to be independent prognostic factors for OS according to multivariate analyses. Kaplan-Meier analysis revealed that CEA and CA24-2 as well as the combination of two were correlated with poor OS. In addition, patients with both markers elevated have worse prognosis than patients with either pre-CEA or post-CA24-2 elevated. Thus, we concluded that the combination of CEA and CA24-2 can be used as a prognostic factor for stage I and II resectable PDAC patients.
Collapse
Affiliation(s)
- Xiaojie Li
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China, 300060
| | - Shengnan Li
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China, 300060
| | - Lili Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Medical University, Tianjin, China, 300060
| | - Jiahui Hong
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Medical University, Tianjin, China, 300060
| | - Tiansuo Zhao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China, 300060
| | - Chuntao Gao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China, 300060
| |
Collapse
|
27
|
Sun Q, Zhang Y, Liu M, Ye Z, Yu X, Xu X, Qin Y. Prognostic and diagnostic significance of galectins in pancreatic cancer: a systematic review and meta-analysis. Cancer Cell Int 2019; 19:309. [PMID: 31832021 PMCID: PMC6873495 DOI: 10.1186/s12935-019-1025-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023] Open
Abstract
Background Galectins constitute a family of β-galactoside-binding proteins, which influence various hallmarks of pancreatic cancer, including cell proliferation, invasion and migration; immune escape; and angiogenesis. Although many studies have concentrated on the role of galectins in pancreatic cancer, the results remain controversial. Hence, we performed a comprehensive meta-analysis to clarify the precise diagnostic and prognostic value of galectins in pancreatic cancer. Methods PubMed/MEDLINE, EMBASE and Web of Science were used to search related published literature up to July 2019. Pooled hazard ratios (HRs), diagnostic accuracy variables and related 95% confidence intervals (CIs) were calculated using STATA 14.0 software. Results Eleven studies including 1227 participants met our inclusion criteria. High expression of galectin family was not correlated with overall survival (OS) in pancreatic cancer (HR, 1.19; 95% CI 0.67-2.11). According to subgroup analysis, high levels of galectin-1 were significantly correlated with worse OS in pancreatic cancer (HR, 4.77; 95% CI 2.47-9.21), while high levels of tandem-repeat galectins (galectin-4 or galectin-9) predicted both better OS (HR, 0.63; 95% CI 0.46-0.86) and disease-free survival (DFS) (HR, 0.63; 95% CI 0.48-0.83). The expression levels of galectin-3 did not directly correlate with prognosis (HR, 0.99; 95% CI 0.40-2.46). The pooled sensitivity, specificity, positive likelihood ratio, and negative likelihood ratios of galectin-3 were 0.64 (95% CI 0.41-0.82), 0.76 (95% CI 0.59-0.88), 2.70 (95% CI 1.21-6.1), and 0.47 (95% CI 0.23-0.98), respectively. The area under the curve (AUC) of galectin-3 was 0.77. Conclusion Taken together, our results suggest that high expression of galectin-1 and low levels of galectin-4 or galectin-9 are predictors of worse prognosis in pancreatic cancer patients. The expression of galectin-3 was not directly related to OS and other clinical characteristics. Although galectin-3 exhibited some diagnostic value in patients with pancreatic cancer in this meta-analysis, clinical application prospects remain to be validated. Further studies are warranted to confirm and strengthen these findings.
Collapse
Affiliation(s)
- Qiqing Sun
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Yiyin Zhang
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Mengqi Liu
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Zeng Ye
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Xianjun Yu
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Xiaowu Xu
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Yi Qin
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| |
Collapse
|
28
|
Du X, Zheng X, Zhang Z, Wu X, Sun L, Zhou J, Liu M. A Label-Free Electrochemical Immunosensor for Detection of the Tumor Marker CA242 Based on Reduced Graphene Oxide-Gold-Palladium Nanocomposite. NANOMATERIALS 2019; 9:nano9091335. [PMID: 31540374 PMCID: PMC6781068 DOI: 10.3390/nano9091335] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/03/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
Abstract
As a tumor marker, carbohydrate antigen 24-2 (CA242) is a highly accurate and specific diagnostic indicator for monitoring pancreatic and colorectal cancers. The goal of this study was to create a novel label-free electrochemical immunosensor using a nanocomposite glassy carbon electrode for the detection of CA242. Graphene oxide (GO) and polyvinyl pyrrolidone were chosen as the dopants for the preparation of a high-performance reduced-GO-gold-palladium (rGO-Au-Pd) nanocomposite. RGO-Au-Pd was characterized using X-ray diffraction and transmission electron microscopy, revealing that the material exhibited superior electrochemical redox activity and electron transfer ability. The effects of the synthesis method, material concentration, reduction cycle, and pH were investigated to optimize the performance of the immunosensor. As a result of the catalytic activity and biocompatibility of rGO-Au-Pd, the prepared CA242 immunosensor displayed a wide linear range of detection from 0.001 U/mL to 10,000 U/mL with a detection limit of 1.54 × 10−3 U/mL and a sensitivity of 4.24 μA (log10CCA242)−1. More importantly, the immunosensor exhibited satisfactory reproducibility and selectivity when detected CA242 in PBS or human serum. The results of our study provide a platform for the development of novel bioassays for use in early cancer diagnosis and promote the application of biosensing technology in the medical field.
Collapse
Affiliation(s)
- Xin Du
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, Shandong, China
| | - Xiaodi Zheng
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, Shandong, China
| | - Zhenhua Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, Shandong, China
| | - Xiaofan Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of the Ministry of Education, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Lei Sun
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, Shandong, China
| | - Jun Zhou
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, Shandong, China
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of the Ministry of Education, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Min Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan 250014, Shandong, China.
| |
Collapse
|
29
|
Tartaglione S, Pecorella I, Zarrillo SR, Granato T, Viggiani V, Manganaro L, Marchese C, Angeloni A, Anastasi E. Protein Induced by Vitamin K Absence II (PIVKA-II) as a potential serological biomarker in pancreatic cancer: a pilot study. Biochem Med (Zagreb) 2019; 29:020707. [PMID: 31223261 PMCID: PMC6559614 DOI: 10.11613/bm.2019.020707] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 03/28/2019] [Indexed: 12/25/2022] Open
Abstract
Introduction Protein induced by vitamin K absence II (PIVKA-II) is an abnormal prothrombin increased in gastrointestinal malignancy. We aimed to evaluate PIVKA-II in comparison to established pancreatic cancer (PC) biomarkers (CA 19-9, carcinoembryonic antigen (CEA) and CA 242) measured in PC patients and in patients with benign pancreatic diseases. Materials and methods We studied 26 PC patients (Group 1) and 20 patients with benign pancreatic diseases (Group 2). PIVKA-II and CEA were measured by chemiluminescent enzyme immunoassay method (CLEIA) on LUMIPULSE G1200 (Fujirebio-Europe, Gent, Belgium), CA 19-9 and CA 242 were measured by ELSA (CisBio Bioassays, Codolet, France) and EIA (Fujirebio Diagnostics AB, Göteborg, Sweden), respectively. Receiver operating characteristic (ROC) analysis was performed to assess biomarkers’ diagnostic characteristics in both groups. Results Median and interquartile range (IQR) in Group 1 and Group 2 were: 1749.0 (320.2 – 3921.0) vs. 31.0 (23.0 – 43.0) mAU/mL (P < 0.001) for PIVKA-II, 260.0 (158.7 – 272.0) vs. 45.2 (9.0 – 58.0) U/mL (P = 0.034) for CA 19-9, 104.0 (30.2 – 150.0) vs. 7.2 (4.8 – 26.0) U/mL (P < 0.050) for CA 242, 9.4 (5.3 – 37.5) vs. 4.5 (1.8 – 7.0) ng/mL (P = 0.021) for CEA. Areas under the ROC curve of PIVKA-II, CA 19-9, CA 242, CEA were 0.86 (95% CI: 0.71 – 1.00), 0.58 (95% CI: 0.38 – 0.78), 0.73 (95% CI: 0.54 – 0.92), 0.64 (95% CI: 0.44 – 0.85), respectively. Conclusions PIVKA-II is significantly higher in PC than in benign pancreatic diseases. PIVKA-II shows a rather good diagnostic performance compared to CA 19-9, CEA and CA242, thus its determination could help PC management.
Collapse
Affiliation(s)
- Sara Tartaglione
- Department of Molecular Medicine, Policlinico Umberto I, University of Rome "Sapienza", Rome, Italy
| | - Irene Pecorella
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Policlinico Umberto I, University of Rome "Sapienza", Rome, Italy
| | - Serena Rita Zarrillo
- Department of Molecular Medicine, Policlinico Umberto I, University of Rome "Sapienza", Rome, Italy
| | | | - Valentina Viggiani
- Department of Molecular Medicine, Policlinico Umberto I, University of Rome "Sapienza", Rome, Italy
| | - Lucia Manganaro
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Policlinico Umberto I, University of Rome "Sapienza", Rome, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, Policlinico Umberto I, University of Rome "Sapienza", Rome, Italy
| | - Antonio Angeloni
- Department of Experimental Medicine, Policlinico Umberto I, University of Rome "Sapienza", Rome, Italy
| | - Emanuela Anastasi
- Department of Molecular Medicine, Policlinico Umberto I, University of Rome "Sapienza", Rome, Italy
| |
Collapse
|
30
|
CA242 as a biomarker for pancreatic cancer and other diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 162:229-239. [PMID: 30905452 DOI: 10.1016/bs.pmbts.2018.12.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
CA242 is a sialic acid-containing carbohydrate antigen attached to core proteins/lipids detected on the cell surface or in serum. Increased serum levels of CA242 have been used clinically as diagnostic biomarkers for pancreatic, colorectal, and other cancers. Since CA242 is overexpressed in malignant tumor tissues compared to that in normal tissues, it is believed that serum CA242 is a product of cancer cells. Thus far, the serum CA242 levels in patients suffering from different types of cancer and noncancerous diseases have not been systematically compared. In our current study, we acquired data of serum CA242 levels from 34,680 patients with 27 clinically defined diseases and from healthy controls (1317) in the clinical laboratory of our hospital over the past 5 years. The mean, median, and -log10p values were calculated. Our data showed that patients with pancreatic cancer, cervical cancer, and lymphoma had the highest median values of serum CA242, which were followed by esophagus cancer, colon cancer, ovarian cancer, type 2 diabetes mellitus, rectal cancer, coronary heart disease, breast cancer, diabetic nephropathy, acute myocardial infarction, and 15 other types of diseases compared to that of healthy controls. In contrast, patients suffering from sequela of brain damage and anemia had statistically lower median values than healthy controls. Based on the -log10p values, the increased serum CA242 levels could be used not only for diagnosis of pancreatic and other cancers, but also for diagnosis of type 2 diabetes mellitus and coronary heart disease, suggesting CA242 might be a systemic malfunction biomarker associated with cancers and other chronic diseases.
Collapse
|
31
|
Hasan S, Jacob R, Manne U, Paluri R. Advances in pancreatic cancer biomarkers. Oncol Rev 2019; 13:410. [PMID: 31044028 PMCID: PMC6478006 DOI: 10.4081/oncol.2019.410] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 01/21/2019] [Indexed: 12/16/2022] Open
Abstract
Biomarkers play an essential role in the management of patients with invasive cancers. Pancreatic ductal adenocarcinoma (PDC) associated with poor prognosis due to advanced presentation and limited therapeutic options. This is further complicated by absence of validated screening and predictive biomarkers for early diagnosis and precision treatments respectively. There is emerging data on biomarkers in pancreatic cancer in past two decades. So far, the CA 19-9 remains the only approved biomarker for diagnosis and response assessment but limited by low sensitivity and specificity. In this article, we aim to review current and future biomarkers that has potential serve as critical tools for early diagnostic, predictive and prognostic indications in pancreatic cancer.
Collapse
Affiliation(s)
- Syed Hasan
- University of Alabama at Birmingham, USA
| | | | | | | |
Collapse
|
32
|
Stoffel EM, McKernin SE, Brand R, Canto M, Goggins M, Moravek C, Nagarajan A, Petersen GM, Simeone DM, Yurgelun M, Khorana AA. Evaluating Susceptibility to Pancreatic Cancer: ASCO Provisional Clinical Opinion. J Clin Oncol 2019; 37:153-164. [DOI: 10.1200/jco.18.01489] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose An ASCO provisional clinical opinion (PCO) offers timely clinical direction to ASCO’s membership and other health care providers. This PCO addresses identification and management of patients and family members with possible predisposition to pancreatic adenocarcinoma. Methods ASCO convened an Expert Panel and conducted a systematic review of the literature published from January 1998 to June 2018. Results of the databases searched were supplemented with hand searching of the bibliographies of systematic reviews and selected seminal articles and contributions from Expert Panel members’ curated files. Provisional Clinical Opinion All patients diagnosed with pancreatic adenocarcinoma should undergo assessment of risk for hereditary syndromes known to be associated with an increased risk for pancreatic adenocarcinoma. Assessment of risk should include a comprehensive review of family history of cancer. Individuals with a family history of pancreatic cancer affecting two first-degree relatives meet criteria for familial pancreatic cancer (FPC). Individuals (cancer affected or unaffected) with a family history of pancreatic cancer meeting criteria for FPC, those with three or more diagnoses of pancreatic cancer in same side of the family, and individuals meeting criteria for other genetic syndromes associated with increased risk for pancreatic cancer have an increased risk for pancreatic cancer and are candidates for genetic testing. Germline genetic testing for cancer susceptibility may be discussed with individuals diagnosed with pancreatic cancer, even if family history is unremarkable. Benefits and limitations of pancreatic cancer screening should be discussed with individuals whose family history meets criteria for FPC and/or genetic susceptibility to pancreatic cancer. Additional information is available at www.asco.org/gastrointestinal-cancer-guidelines .
Collapse
Affiliation(s)
| | | | | | | | | | | | - Arun Nagarajan
- Taussig Cancer Institute and Case Comprehensive Cancer Center, Cleveland Clinic, Cleveland, OH
| | | | | | | | - Alok A. Khorana
- Taussig Cancer Institute and Case Comprehensive Cancer Center, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
33
|
Peng HY, Chang MC, Hu CM, Yang HI, Lee WH, Chang YT. Thrombospondin-2 is a Highly Specific Diagnostic Marker and is Associated with Prognosis in Pancreatic Cancer. Ann Surg Oncol 2018; 26:807-814. [PMID: 30569296 DOI: 10.1245/s10434-018-07109-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Thrombospondin-2 (TSP-2) has been reported as an early diagnostic marker for pancreatic ductal adenocarcinoma (PDAC) in Caucasian populations. This study was designed to validateTSP-2 as a diagnostic marker in a large Taiwan cohort and to investigate the association of TSP-2 with the clinical outcomes of PDAC patients. METHODS The serum TSP-2 levels in 263 PDAC patients and 230 high-risk individuals (HRIs) were measured via an enzyme-linked immunosorbent assay. The sensitivity, specificity, and accuracy of TSP-2 as a diagnostic marker to discriminating PDAC patients from HRIs and correlations between TSP-2 levels and prognosis of PDAC patients were analyzed. RESULTS Serum TSP-2 levels were significantly higher in patients with PDAC (44.90 ± 40.70 ng/ml) than in the HRIs (17.52 ± 6.23 ng/ml). At a level of ≥ 29.8 ng/ml, TSP-2 exhibited 100% specificity, 55.9% sensitivity, 100% positive predictive value (PPV), and 66.5% negative predictive value (NPV) for discriminating PDAC patients from HRIs. The Cox regression analysis showed that higher serum TSP-2 levels were significantly associated with poor outcomes in PDAC patients (hazard ratio = 1.54, 95% confidence interval = 1.143-2.086, P = 0.005). Combining the carbohydrate antigen 19-9 (CA19-9) (cutoff value of 62.0 U/ml) and TSP-2 (cutoff value of 29.8 ng/ml) levels yielded 98.7% specificity, 90.5% sensitivity, 98.8% PPV, and 90.1% NPV for discriminating patients with PDAC from HRIs. CONCLUSIONS TSP-2 is a highly specific diagnostic marker and an independent prognostic marker in patients with PDAC. A combined biomarker panel, including TSP-2 and CA19-9, may facilitate future PDAC screening.
Collapse
Affiliation(s)
- Hsuan-Yu Peng
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Chu Chang
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Mei Hu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hwai-I Yang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wen-Hwa Lee
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Taiwan Graduate Institute of Clinical Medicine, China Medical University, Taichung, Taiwan
| | - Yu-Ting Chang
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan. .,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
34
|
Nora I, Shridhar R, Huston J, Meredith K. The accuracy of neutrophil to lymphocyte ratio and platelet to lymphocyte ratio as a marker for gastrointestinal malignancies. J Gastrointest Oncol 2018; 9:972-978. [PMID: 30505600 DOI: 10.21037/jgo.2018.08.05] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Accurate predictors of locally advanced and recurrence disease in patients with gastrointestinal cancer are currently lacking. Neutrophil to lymphocyte ratio (NLR) and platelet to lymphocyte ratio (PLR) have emerged as possible markers for predicting recurrence in these patients. In this study, we sought to evaluate the utility of NLR and PLR in predicting the presence of regional nodal disease, metastasis and systemic recurrence in patients with gastrointestinal malignancies. Methods We queried a comprehensive gastrointestinal oncology database to identify patients who had undergone surgery for a GI malignancy. NLR and PLR values were determined via a complete blood count (CBC). In patients treated with neoadjuvant therapy (NT) the NLR and PLR were calculated from CBCs before and after NT and in patients proceeding to surgery within 2 weeks pre-operatively. The associations between NLR and PLR and the clinicopathologic parameters (sex, age, tumor size, differentiation, positive lymph nodes, and metastatic disease) were assessed via χ2 or Fisher's exact tests where appropriate. All the tests were two-sided, and P<0.05 was considered statistically significant. Results We identified 116 patients diagnosed with gastrointestinal malignancies. There were 76 (65.5%) males and 40 (34.5%) females with an average age of 69.4±10.7 years. The mean follow up was 14.1±15.5 months. We identified 49 (42.2%) esophageal, 34 (29.3%) pancreatic, 14 (12.1%) colorectal, 13 (11.2%) gastric, and 6 (5.2%) biliary cancers. There were 36 (31.0%) patients with node negative disease, 52 (44.8%) with node positive and 28 (24.2%) with metastatic disease at surgery. Of the metastatic patients 4 (3.4%) were found at staging laparoscopy and 24 (20.6%) were diagnosed pre-operatively. The median NLR for LN- patient's was 1.78 (0.23-8.2) and for LN+ and metastatic patients was 4.69 (2.27-36), P<0.001. The median PLR for LN- patient's was 123.03 (14-257.69) and for LN+ and metastatic patients was 212.42 (105.45-2,185.18), P<0.001. The sensitivity, specificity, positive predictive value (PPV) and negative predictive value (NPV) for a NLR >2.25 was 98.8%, 72.2%, 89%, and 96% respectively. The sensitivity, specificity, PPV, and NPV for PLR >140 was 95%, 78%, 90%, and 88% respectively. Utilizing both NLR and PLR the sensitivity, specificity, PPV and NPV was increased. Conclusions Elevation of NLR and PLR can be used to help identify patients with advanced disease GI malignancies and recurrences after surgery. Additionally, failure of normalization of NLR and PLR 3-month post-surgical resection may indicate early recurrence or persistent disease. Individually, NLR has a higher sensitivity and negative predictive value while PLR has a higher specificity and positive predictive value for distinguishing metastatic disease and node positivity. The combination of NLR and PLR has the highest accuracy of predicting advanced disease among all gastrointestinal malignancies.
Collapse
Affiliation(s)
- Ian Nora
- College of Medicine, University of Limerick, Limerick, Ireland
| | - Ravi Shridhar
- Department of Radiation Oncology, Florida Hospital Cancer Institute, Orlando, FL, USA
| | - Jamie Huston
- Department of Gastrointestinal Oncology, Sarasota Memorial Healthcare System, Sarasota, FL, USA
| | - Kenneth Meredith
- Department of Gastrointestinal Oncology, Sarasota Memorial Healthcare System, Sarasota, FL, USA.,Department of Gastrointestinal Oncology, Florida State University College of Medicine, Tallahassee, FL, USA
| |
Collapse
|
35
|
Pang L, Zhang N, Xia Y, Wang D, Wang G, Meng X. Serum APN/CD13 as a novel diagnostic and prognostic biomarker of pancreatic cancer. Oncotarget 2018; 7:77854-77864. [PMID: 27788483 PMCID: PMC5363626 DOI: 10.18632/oncotarget.12835] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 10/14/2016] [Indexed: 12/24/2022] Open
Abstract
Aminopeptidase N, also known as CD13, has been reported to be overexpressed in several cancers and may contribute to tumor metastasis and angiogenesis. The aim of this study was to evaluate whether serum APN/CD13 could be a potential biomarker for the diagnosis and prognosis of pancreatic cancer (PC). Serum APN/CD13 and carbohydrate antigen 19-9 (CA19-9) levels were measured from 382 participants, which comprised of 204 participants with PC, 48 participants with benign pancreatic tumors (BPT), 43 participants with chronic pancreatitis (CP) and 87 healthy controls (HC). We used receiver operating characteristic (ROC) analysis to calculate diagnostic accuracy. The association of serum APN/CD13 levels with the clinicopathological characteristics of PC patients and their survival was investigated. Serum APN/CD13 levels were substantially higher in PC patients than in controls. ROC analysis revealed that APN/CD13 was significantly better than CA19-9 in differentiating patients with PC from controls. Similar results were noted for early-stage PC. Moreover, the combined use of APN/CD13 and CA19-9 data improved the diagnostic accuracy for PC vs. controls, compared with either test alone. High serum APN/CD13 levels were associated with tumor size, lymph nodes, and metastasis (TNM) stage. Multivariate and ROC curve analyses revealed that high serum APN/CD13 level is an independent factor for predicting mortality and overall survival (OS). Moreover, Kaplan–Meier analysis demonstrated an inverse correlation between increased serum APN/CD13 level and OS. Our study established that serum APN/CD13 may be a novel diagnostic and prognostic biomarker for PC.
Collapse
Affiliation(s)
- Li Pang
- Department of Emergency, The First Hospital of Jilin University, Changchun 130021, China
| | - Nan Zhang
- Department of Emergency, The First Hospital of Jilin University, Changchun 130021, China
| | - Yan Xia
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun 130021, China
| | - Dawei Wang
- Department of Emergency, The First Hospital of Jilin University, Changchun 130021, China
| | - Guoqing Wang
- Basic Medical College of Jilin University, Changchun 130021, China
| | - Xiangwei Meng
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
36
|
Mayerle J, Kalthoff H, Reszka R, Kamlage B, Peter E, Schniewind B, González Maldonado S, Pilarsky C, Heidecke CD, Schatz P, Distler M, Scheiber JA, Mahajan UM, Weiss FU, Grützmann R, Lerch MM. Metabolic biomarker signature to differentiate pancreatic ductal adenocarcinoma from chronic pancreatitis. Gut 2018; 67:128-137. [PMID: 28108468 PMCID: PMC5754849 DOI: 10.1136/gutjnl-2016-312432] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 12/22/2016] [Accepted: 12/26/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Current non-invasive diagnostic tests can distinguish between pancreatic cancer (pancreatic ductal adenocarcinoma (PDAC)) and chronic pancreatitis (CP) in only about two thirds of patients. We have searched for blood-derived metabolite biomarkers for this diagnostic purpose. DESIGN For a case-control study in three tertiary referral centres, 914 subjects were prospectively recruited with PDAC (n=271), CP (n=282), liver cirrhosis (n=100) or healthy as well as non-pancreatic disease controls (n=261) in three consecutive studies. Metabolomic profiles of plasma and serum samples were generated from 477 metabolites identified by gas chromatography-mass spectrometry and liquid chromatography-tandem mass spectrometry. RESULTS A biomarker signature (nine metabolites and additionally CA19-9) was identified for the differential diagnosis between PDAC and CP. The biomarker signature distinguished PDAC from CP in the training set with an area under the curve (AUC) of 0.96 (95% CI 0.93-0.98). The biomarker signature cut-off of 0.384 at 85% fixed specificity showed a sensitivity of 94.9% (95% CI 87.0%-97.0%). In the test set, an AUC of 0.94 (95% CI 0.91-0.97) and, using the same cut-off, a sensitivity of 89.9% (95% CI 81.0%-95.5%) and a specificity of 91.3% (95% CI 82.8%-96.4%) were achieved, successfully validating the biomarker signature. CONCLUSIONS In patients with CP with an increased risk for pancreatic cancer (cumulative incidence 1.95%), the performance of this biomarker signature results in a negative predictive value of 99.9% (95% CI 99.7%-99.9%) (training set) and 99.8% (95% CI 99.6%-99.9%) (test set). In one third of our patients, the clinical use of this biomarker signature would have improved diagnosis and treatment stratification in comparison to CA19-9.
Collapse
Affiliation(s)
- Julia Mayerle
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany,Medizinische Klinik und Poliklinik II, Klinikum der LMU München-Grosshadern, München, Germany
| | - Holger Kalthoff
- Section for Molecular Oncology, Institut for Experimental Cancer Research (IET), UKSH, Kiel, Germany
| | | | | | | | - Bodo Schniewind
- Section for Molecular Oncology, Institut for Experimental Cancer Research (IET), UKSH, Kiel, Germany
| | | | | | - Claus-Dieter Heidecke
- Department of General, Visceral, Thoracic and Vascular Surgery University Medicine Greifswald, Ernst-Moritz-Arndt University, Greifswald, Germany
| | | | - Marius Distler
- Clinic and Outpatient Clinic for Visceral-, Thorax- and Vascular Surgery, Medizinische Fakultät, TU Dresden, Dresden, Germany
| | - Jonas A Scheiber
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Ujjwal M Mahajan
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany,Medizinische Klinik und Poliklinik II, Klinikum der LMU München-Grosshadern, München, Germany
| | - F Ulrich Weiss
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | | | - Markus M Lerch
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| |
Collapse
|
37
|
Zhu SK. Role of precision medicine in pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2016; 24:4752-4758. [DOI: 10.11569/wcjd.v24.i36.4752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the most challenging problems in modern oncology. Due to difficultly in early diagnosis and early distant metastasis of pancreatic cancer, surgical resection rate is less than 20% and patients' prognosis is very poor. Despite long-term efforts taken to develop treatments for pancreatic cancer, the survival rate did not significantly improve. Therefore, early diagnosis and treatment are the key to improve the survival rate of patients with pancreatic cancer. The advent of big-data genomic era and the rapid development of biotechnology have led to the recent proposal of a new concept of precise medicine, which has quickly become the focus of world medical conferences. Here, we describe the new progress and challenges of precision medicine in pancreatic cancer, with an aim to provide new ideas for improving the survival rate of patients with pancreatic cancer.
Collapse
|
38
|
Serum CA242, CA199, CA125, CEA, and TSGF are Biomarkers for the Efficacy and Prognosis of Cryoablation in Pancreatic Cancer Patients. Cell Biochem Biophys 2016; 71:1287-91. [PMID: 25486903 DOI: 10.1007/s12013-014-0345-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study aimed to compare the changes and determine the clinical significance of carbohydrate antigens CA242, CA199, CA125, carcinoembryonic antigen (CEA), and tumor-specific growth factor (TSGF) before and after cryoablation by Cryocare system. Thirty-one pancreatic cancer patients were selected to receive cryoablation by Cryocare system. The serum expression levels of CA242, CA199, CA125, CEA, and TSGF before and 1 month after treatment were determined. Meanwhile, the serum level of these factors was also determined in 31 healthy volunteers. The parameter changes were analyzed with the clinical pathological data. The serum levels of CA242, CA199, CA125, CEA, and TSGF in the pancreatic cancer group were significantly higher than those of the control group both before and after the cryoablation treatment (P < 0.05). The serum CA199, CEA, and TSGF dramatically decreased 1 month after the treatment, which were statistically different (P < 0.05). The positive rates of serum CA242, CA199, CA125, and CEA in the pancreatic cancer group were much higher than those in the control group both before and after treatment (P < 0.05), and the positive rate of TSGF was significantly higher than that of the control group before the treatment (P < 0.05). The positive rate of CA199, CEA, and TSGF after the treatment was significantly lower than that before the treatment (P < 0.05). Serum level of CA242 was correlated with the tumor diameter, clinical staging, tumor differentiation, lymph node, and liver metastasis (P < 0.05). Except gender, CA199 was correlated with all the other clinical pathological parameters (P < 0.05). The serum levels of CA125 and CEA were correlated with all the other clinical pathological parameters (P < 0.05). The serum level of TSGF was only correlated with tumor differentiation (P < 0.05). Cryoablation treatment by Cryocare system can decrease the serum levels of CA199, CEA, TSGF, and the positive rate. Serum CA199, CEA, and TSGF can be important index for pancreatic cancer treatment assessment. Serum levels of CA242, CA199, CA125, and CEA are of great clinical value for metastasis assessment and prognosis in pancreatic cancer patients.
Collapse
|
39
|
Zhao S, Chen X, Wan M, Jiang X, Li C, Cui Y, Kang P. Tectonic 1 Is a Key Regulator of Cell Proliferation in Pancreatic Cancer. Cancer Biother Radiopharm 2016; 31:7-13. [PMID: 26844847 DOI: 10.1089/cbr.2014.1778] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Pancreatic cancer is notoriously becoming one of the most devastating human cancers leading to death. However, clinical challenges still remain in diagnosis and treatment of this ticklish cancer. In the present study, the authors identified a new gene, Tectonic 1 (TCTN1), as a key regulator of cell proliferation in pancreatic cancer. Lentivirus-mediated short hairpin RNA (shRNA) was employed to knock down endogenous TCTN1 expression in PANC-1 pancreatic cancer cells. Knockdown of TCTN1 expression potently inhibited cell viability and proliferation, as determined by MTT and colony formation assays. Western blotting analysis also showed that knockdown of TCTN1 suppressed the expression of cdc2, while it induced that of p21 and p27. Flow cytometry analysis showed that depletion of TCTN1 in PANC-1 cells led to cell cycle arrest in the G2/M phase as well as apoptosis. Besides, depletion of TCTN1 led to the increase of Bax and cleavage of PARP-1, but the decrease of bcl2 by western blotting. The data indicate that TCTN1 is indispensable for pancreatic cancer cell proliferation, which provides a novel alternative to targeted therapy of pancreatic cancer and deserves further investigation.
Collapse
Affiliation(s)
- Shiyong Zhao
- 1 Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University , Harbin, China
| | - Xuedong Chen
- 2 Department of Ophthalmology, the First Affiliated Hospital of Harbin Medical University , Harbin, China
| | - Ming Wan
- 3 Harbin Medical University , Harbin, China
| | - Xingming Jiang
- 1 Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University , Harbin, China
| | - Chunlong Li
- 1 Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University , Harbin, China
| | - Yunfu Cui
- 1 Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University , Harbin, China
| | - Pengcheng Kang
- 1 Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University , Harbin, China
| |
Collapse
|
40
|
Lerch MM, Mayerle J, Mahajan U, Sendler M, Weiss FU, Aghdassi A, Moskwa P, Simon P. Development of Pancreatic Cancer: Targets for Early Detection and Treatment. Dig Dis 2016; 34:525-31. [PMID: 27332960 DOI: 10.1159/000445233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is the 4th leading cause of cancer death worldwide and compared to other malignancies its share in cancer mortality is expected to rise further. This is due to a lack of sensitive diagnostic tools that would permit earlier detection in a potentially curable stage and the very slow progress in finding effective drug treatments for pancreatic cancer. KEY MESSAGES Aside from genetic predispositions and environmental agents, chronic pancreatitis is by far the greatest risk factor for PDAC. It also shares several etiological factors with pancreatic cancer and represents its most challenging differential diagnosis. Biomarkers that can distinguish between chronic pancreatitis and PDAC may therefore be suitable for the latter's early detection. Moreover, targeting the natural history of chronic pancreatitis would be one approach to prevent PDAC. Targeting tumor-cell signaling directly by interfering with receptor tyrosine kinases has shown some efficacy, although the results in clinical trials were less encouraging than for other cancers. Other compounds developed have targeted the formation of extracellular matrix around the tumor, the proteolytic activity in the tumor environment, histone deacetylases, hedgehog signaling and heat shock proteins, but none has yet found its way into routine patient care. Attempts to individualize treatment according to the tumor's somatic mutation profile are novel but so far impractical. CONCLUSIONS Progress in the treatment of pancreatic cancer has been exceedingly slow and mostly dependent on improved pharmaceutical preparations or combinations of established chemotherapeutic agents. The promise of major breakthroughs implied in targeting tumor signal transduction events has so far not materialized.
Collapse
Affiliation(s)
- Markus M Lerch
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Yuan C, Yang K, Tang H, Chen D. Diagnostic values of serum tumor markers Cyfra21-1, SCCAg, ferritin, CEA, CA19-9, and AFP in oral/oropharyngeal squamous cell carcinoma. Onco Targets Ther 2016; 9:3381-6. [PMID: 27350753 PMCID: PMC4902246 DOI: 10.2147/ott.s105672] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background At present, the research on serum tumor markers in the early diagnosis of malignant tumors has aroused widespread concern. The aim of this study was to investigate the diagnostic values of serum tumor markers cytokeratin 19 fragment (Cyfra21-1), squamous cell carcinoma antigen (SCCAg), ferritin, carcinoembryonic antigen (CEA), carbohydrate antigen 19-9 (CA19-9), and α-fetoprotein (AFP) for patients with oral/oropharyngeal squamous carcinoma (OSCC/OPSCC). Methods One hundred and sixty-nine cases of patients with OSCC/OPSCC as the experimental group, 86 cases of oral benign tumor patients as the control group, and 30 cases of healthy people as the normal control group were studied. The levels of serum Cyfra21-1, SCCAg, ferritin, CEA, CA19-9, and AFP were measured using electrochemiluminescence immunoassay. Results The levels of serum Cyfra21-1, SCCAg, ferritin, and CEA in patients with OSCC/OPSCC were significantly higher than those of benign tumor and healthy control group (P<0.05). The levels of CA19-9 and AFP showed no significant difference between patients with OSCC/OPSCC, benign tumor, and healthy group (P>0.05). The level of serum Cyfra21-1 in patients with early OSCC/OPSCC (stage I + II) was significantly higher than that of benign tumor and healthy control group (P<0.05). However, the levels of serum SCCAg, ferritin, CEA, CA19-9, and AFP showed no significant difference between patients with early OSCC/OPSCC, benign tumor, and healthy control group (P>0.05). The levels of serum Cyfra21-1, SCCAg, ferritin, and CEA in the middle-late stage of patients with OSCC/OPSCC (stage III + IV) were significantly higher than those of patients with the early OSCC/OPSCC, benign tumor, and healthy control group (P<0.05). The diagnostic cutoff levels of Cyfra21-1, SCCAg, ferritin, and CEA were 2.17, 0.72, 109.95, and 1.99 ng/mL, respectively. The sensitivities were 60.36%, 73.37%, 81.66%, and 66.27%, respectively. The specificities were 81.03%, 68.10%, 40.52%, and 61.21%, respectively. Conclusion Cyfra21-1, SCCAg, ferritin, and CEA had diagnostic values for patients with OSCC/OPSCC. Meanwhile, Cyfra21-1 had better early diagnostic value for patients with OSCC/OPSCC.
Collapse
Affiliation(s)
- Chuanshu Yuan
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Kai Yang
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Hong Tang
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Dan Chen
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
42
|
Gu H, Xin X, Pan Y, Zhang H, Tian S, Sun C. Telomerase activity as a marker for differential diagnosis of pancreatic adenocarcinoma: a systematic review and meta-analysis. Int J Biol Markers 2016; 31:e126-37. [PMID: 26616232 DOI: 10.5301/jbm.5000172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND Studies evaluating the role of telomerase activity in pancreatic adenocarcinoma are inconsistent and a systemic review of the available literature may shed new light on this issue. OBJECTIVE To systematically review the usefulness of telomerase activity in distinguishing pancreatic cancer from other pancreatic diseases. METHODS A comprehensive search of the PubMed and Embase databases was conducted to identify eligible studies. Only studies evaluating telomerase activity in patients with suspected or previously diagnosed pancreatic adenocarcinomas versus nonpancreatic adenocarcinomas and published in English with a sufficient number of cases were included. The hierarchical summary receiver operating characteristic (HSROC) model was used to establish the potential value of telomerase activity in the diagnosis of pancreatic adenocarcinoma. RESULTS A total of 19 studies qualified for this meta-analysis. In distinguishing pancreatic adenocarcinoma from benign diseases, the pooled sensitivity and specificity of telomerase activity were 0.81 (95% CI, 0.68-0.90) and 0.97 (95% CI, 0.93-0.98), respectively; the diagnostic odds ratio (DOR) was 126.62 (95% CI, 49.94-320.99); beta was -1.16 (95% CI, -3.62-1.29), Z was -0.93, p was 0.35>0.1, and lambda was 6.86 (95% CI, 1.01-12.70). In distinguishing pancreatic adenocarcinoma from chronic pancreatitis, the pooled sensitivity and specificity of telomerase activity were 0.77 (95% CI, 0.61-0.88) and 0.97 (95% CI, 0.91-0.99), respectively; DOR was 117.28 (95% CI, 32.25-426.53); beta was -0.38 (95% CI, -1.89-1.13), Z was -0.49, p was 0.62>0.1, and lambda was 5.30 (95% CI, 3.37-7.24). CONCLUSIONS The present meta-analysis demonstrates that telomerase activity could be a useful biomarker for the differential diagnosis of pancreatic adenocarcinoma and benign pancreatic diseases.
Collapse
Affiliation(s)
- Huajian Gu
- Department of General Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang - PR China
| | - Xiaoyan Xin
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan - PR China
| | - Yaozhen Pan
- Department of General Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang - PR China
| | - Hong Zhang
- Department of General Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang - PR China
| | - She Tian
- Department of General Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang - PR China
| | - Chengyi Sun
- Department of General Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang - PR China
| |
Collapse
|
43
|
Han SX, Zhou X, Sui X, He CC, Cai MJ, Ma JL, Zhang YY, Zhou CY, Ma CX, Varela-Ramirez A, Zhu Q. Serum dickkopf-1 is a novel serological biomarker for the diagnosis and prognosis of pancreatic cancer. Oncotarget 2016; 6:19907-17. [PMID: 26101916 PMCID: PMC4637329 DOI: 10.18632/oncotarget.4529] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 05/20/2015] [Indexed: 12/19/2022] Open
Abstract
Purpose To identify whether Dickkopf-1 (DKK1) could be a potential biomarker for early detection and prognosis in patients with pancreatic cancer (PC). Methods Serum was collected from 140 patients with pancreatic adenocarcinoma and 92 control patients without pancreatic adenocarcinoma. Serological levels of DKK1 were examined by enzyme-linked immunosorbent assay (ELISA). The sensitivity and specificity was compared with carbohydrate antigen 19-9 (CA19-9). A 2-year follow-up was monitored to evaluate the correlation between DKK1 serum levels and overall survival. The expression of DKK1 in PC tumor tissues was also evaluated using immunohistochemistry staining. Results Serum levels of DKK1 and CA19-9 were elevated in PC patients in the early-stage cases. These levels increased with the advancement of clinical stage. There was significant difference in DKK1 serum levels between early and advanced PC stages. Receiver operating characteristic curve (ROCC) analysis showed that DKK1 was significantly better than CA19-9 in differentiating patients with PC from the controls (area under the curve (AUC) 0.919 versus 0.853, respectively), especially in distinguishing early-stage cancer from chronic pancreatitis (CP). The expression of DKK1 in PC tissues correlated with its expression in serum samples. The overall survival rate was 24.4% in the group with higher DKK1 levels and was found to be significantly different from the group with lower DKK1 levels (33.3%). Conclusion DKK1 may be a novel diagnostic/prognostic biomarker for PC.
Collapse
Affiliation(s)
- Su-xia Han
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University Medical College, Xi'an, Shaanxi, PR China
| | - Xia Zhou
- Department of Biotherapy, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Xin Sui
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University Medical College, Xi'an, Shaanxi, PR China
| | - Chen-chen He
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University Medical College, Xi'an, Shaanxi, PR China
| | - Meng-jiao Cai
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University Medical College, Xi'an, Shaanxi, PR China
| | - Jin-lu Ma
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University Medical College, Xi'an, Shaanxi, PR China
| | - Yuan-yuan Zhang
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University Medical College, Xi'an, Shaanxi, PR China
| | - Cong-ya Zhou
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University Medical College, Xi'an, Shaanxi, PR China
| | - Chen-xian Ma
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University Medical College, Xi'an, Shaanxi, PR China
| | - Armando Varela-Ramirez
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, USA
| | - Qing Zhu
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University Medical College, Xi'an, Shaanxi, PR China
| |
Collapse
|
44
|
Chronic Pancreatitis: A Review. Indian J Surg 2016; 77:1348-58. [PMID: 27011563 DOI: 10.1007/s12262-015-1221-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 01/05/2015] [Indexed: 10/23/2022] Open
Abstract
This is to incorporate the recent trends in chronic pancreatitis. Extensive literature search was done from Pubmed and Ovid SP. Full text articles and abstracts related to chronic pancreatitis were reviewed and the article was prepared. Chronic pancreatitis is evolving fast on its etiology and treatment areas. The main etiological factors are pointing towards genetic, alcohol, and smoking. Autoimmune has also been added as a cause. Endoscopic ultra sound (EUS) becoming the standard diagnostic procedure. Surgery is becoming the treatment of choice for pain and mechanical complications and not the endotherapy in the long-term follow-up. The numbers of surgeries are getting narrowed down. The two etiological groups of chronic pancreatitis namely alcohol + smoking and genetic getting clearer, where the later group present at an early age. Endoscopic ultrasound and imaging with secretin is diagnostic of chronic pancreatitis before the structural changes. Endotherapy is found to be inferior to surgery in long-term pain relief. Diagnostic criteria for autoimmune pancreatitis are established. Pancreaticogenic diabetes (Type3c) and its problem associated with fat malabsorption are being understood.
Collapse
|
45
|
Ultrasensitive photoelectrochemical immunoassay for CA19-9 detection based on CdSe@ZnS quantum dots sensitized TiO 2 NWs/Au hybrid structure amplified by quenching effect of Ab 2 @V 2+ conjugates. Biosens Bioelectron 2016; 77:339-46. [DOI: 10.1016/j.bios.2015.09.051] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 09/02/2015] [Accepted: 09/23/2015] [Indexed: 12/26/2022]
|
46
|
You L, Ma L, Zhao W, Zhao Y, Dai M. Emerging role of tumor markers and biochemistry in the preoperative invasive assessment of intraductal papillary mucinous neoplasm of the pancreas. Clin Chim Acta 2016; 454:89-93. [DOI: 10.1016/j.cca.2015.12.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 12/09/2015] [Accepted: 12/30/2015] [Indexed: 01/06/2023]
|
47
|
Seufferlein T, Mayerle J. Pancreatic cancer in 2015: Precision medicine in pancreatic cancer--fact or fiction? Nat Rev Gastroenterol Hepatol 2016; 13:74-5. [PMID: 26758788 DOI: 10.1038/nrgastro.2015.215] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Late diagnosis and an inability to personalize treatment are major problems preventing reductions in pancreatic cancer mortality. In 2015, the identification of a highly discriminatory exosomal biomarker, culture systems that recapitulate human disease and new methods of analysing large data sets to identify prognostic markers have improved the future outlook for patients with this cancer.
Collapse
Affiliation(s)
- Thomas Seufferlein
- Ulm University Medical Center, Department of Internal Medicine I, Albert Einstein Allee 23, D-89081 Ulm, Germany
| | - Julia Mayerle
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Ferdinand-Sauerbruchstrasse, 17475 Greifswald, Germany
| |
Collapse
|
48
|
Jiang XH, Hu NZ, Wei MT. Value of 18F-fluorodeoxyglucose positron emission tomography and 18F-fluorodeoxyglucose positron emission tomography/computed tomography in diagnosis of pancreatic cancer: A systemic review and meta-analysis. Shijie Huaren Xiaohua Zazhi 2016; 24:136-146. [DOI: 10.11569/wcjd.v24.i1.136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To evaluate the value of 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) and 18F-FDG PET/computed tomography (CT) in the diagnosis of pancreatic cancer.
METHODS: Medline, EMBASE, Science Direct, Springer link, CBM, Cnki, Wan fang and VIP databases were searched by computer before April 1, 2015 to retrieve articles on the study of 18F-FDG PET and 18F-FDG PET/CT in diagnosing pancreatic cancer. Studies were selected according to the inclusion and exclusion criteria, and quality assessment was made using the QUADAS scale. Meta-Disc 1.4 software was used to analyze the heterogeneity of the included articles, and the SROC curve was plotted to calculate the pooled sensitivity and specificity. The publication bias was assessed with Stata 12.0 software.
RESULTS: A total of 51 English-language articles were included. The summary sensitivity and specificity of 18F-FDG PET in diagnosing pancreatic cancer were 87% (95%CI: 85%-89%) and 78% (95%CI: 74%-81%), respectively. The positive and negative likelihood ratios were 3.38 (95%CI: 2.64-4.33) and 0.18 (95%CI: 0.14-0.23), respectively. The diagnostic odds ratio (DOR) was 21.91 (95%CI: 14.15-33.93), and the area under the SROC curve was 0.8930. The summary sensitivity and specificity of 18F-FDG PET/CT in diagnosing pancreatic cancer were 91% (95%CI: 88%-93%) and 77% (95%CI: 72%-82%), respectively. The positive and negative likelihood ratios were 3.57 (95%CI: 2.96-4.31) and 0.14 (95%CI: 0.11-0.18), respectively. The DOR was 28.52 (95%CI: 19.63-41.42), and the area under the SROC curve was 0.9315.
CONCLUSION: 18F-FDG PET/CT and 18F-FDG PET have higher diagnostic value than CT in diagnosing pancreatic cancer. 18F-FDG PET/CT is superior to 18F-FDG PET in terms of sensitivity and both of them can be used as diagnostic tools for pancreatic cancer with negative traditional examinations.
Collapse
|
49
|
Rong Q, Feng F, Ma Z. Metal ions doped chitosan–poly(acrylic acid) nanospheres: Synthesis and their application in simultaneously electrochemical detection of four markers of pancreatic cancer. Biosens Bioelectron 2016; 75:148-54. [DOI: 10.1016/j.bios.2015.08.041] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 08/09/2015] [Accepted: 08/19/2015] [Indexed: 11/25/2022]
|
50
|
Zhang A, Xiang H, Zhang X, Guo W, Yuan E, Huang C, Jia N. A novel sandwich electrochemiluminescence immunosensor for ultrasensitive detection of carbohydrate antigen 19-9 based on immobilizing luminol on Ag@BSA core/shell microspheres. Biosens Bioelectron 2016; 75:206-12. [DOI: 10.1016/j.bios.2015.08.047] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 08/10/2015] [Accepted: 08/21/2015] [Indexed: 12/24/2022]
|