1
|
Tan AYS, Cheng F, Zhang M, Tan MTT, Manickam S, Muthoosamy K. Graphitic carbon nitride/1-pyrenebutyric acid N-hydroxysuccinimide/polythiophene nanocomposite photoelectrochemical biosensor for CA 19-9 detection. Talanta 2025; 293:128065. [PMID: 40253965 DOI: 10.1016/j.talanta.2025.128065] [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: 11/03/2024] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/22/2025]
Abstract
A photoelectrochemical (PEC) biosensor composed entirely of carbon nanomaterials was synthesized to detect carbohydrate antigen 19-9 (CA 19-9). The biosensor platform integrated graphitic carbon nitride (GCN), known for its light sensitivity, polythiophene (PTh), an organic conductive and optically active material, and 1-pyrenebutyric acid N-hydroxysuccinimide (PBASE), which functions both as a biolinker to conjugate CA 19-9 antibody and antigen and as an electron mediator to facilitate electron transfer from GCN to PTh. The formation of a Schottky heterojunction between PTh and GCN reduced the bandgap of GCN from 2.66 to 1.96 eV, which enhanced transfer of photogenerated electrons for cathodic photocurrent generation. The improvement of charge transfer due to heterojunction formation and π-π stacking between GCN and the pyrene group of PBASE is confirmed by cyclic voltammetry (CV), electron impedance spectroscopy (EIS), and chronoamperometry (CA) findings. The highest current of 1.31 μA is observed for combination of 5 wt% PTh with a GCN/PBASE ratio of 1:0.5. Besides evaluating the electron mobility of GCN/PBASE/PTh, CV, EIS, and CA were also used to evaluate the sensor performance. Optimization studies revealed that 0.6 μg of CA 19-9 antibody and 1 h of antigen-antibody immobilization time significantly improved the biosensor response. The GCN/PBASE/PTh biosensor demonstrated high sensitivity to CA 19-9 antigen across a concentration of 50-1000 U/ml and a detection limit as low as 0.052 U/ml. The reported working range is within the limits required for diagnostic testing of patients with hepatic and heart problems as well as for post-treatment monitoring of colorectal and pancreatic cancer patients.
Collapse
Affiliation(s)
- Adriel Yan Sheng Tan
- Guangdong Engineering and Technology Research Centre for Advanced Nanomaterials, School of Environment and Civil Engineering, Dongguan University of Technology, Dongguan, 523808, China; Nanotechnology Research Group, Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, 43500 Semenyih, Selangor, Malaysia
| | - Faliang Cheng
- Guangdong Engineering and Technology Research Centre for Advanced Nanomaterials, School of Environment and Civil Engineering, Dongguan University of Technology, Dongguan, 523808, China.
| | - Min Zhang
- Guangdong Engineering and Technology Research Centre for Advanced Nanomaterials, School of Environment and Civil Engineering, Dongguan University of Technology, Dongguan, 523808, China
| | - Michelle T T Tan
- Department of Electrical and Electronic Engineering, Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, 43500 Semenyih, Selangor, Malaysia
| | - Sivakumar Manickam
- Petroleum and Chemical Engineering, Faculty of Engineering, Universiti Teknologi Brunei, Bandar Seri Begawan, BE1410, Brunei Darussalam
| | - Kasturi Muthoosamy
- Nanotechnology Research Group, Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, 43500 Semenyih, Selangor, Malaysia.
| |
Collapse
|
2
|
Liu C, Mosley A, Irajizad E, Yip-Schneider M, Wu H, Smith-Kinnaman WR, Tran T, Long JP, Do KA, Fahrmann J, DeWitt JM, Hanash S, Schmidt CM, Zhang J. Cyst fluid proteins stratify malignant risk of intraductal papillary mucinous neoplasm of the pancreas. Cancer Lett 2025; 624:217753. [PMID: 40300662 DOI: 10.1016/j.canlet.2025.217753] [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: 01/08/2025] [Revised: 04/02/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
Intraductal papillary mucinous neoplasm (IPMN) is the most common type of pancreatic cyst often incidentally detected in asymptomatic patients. The current consensus guidelines, largely based on imaging features, have high sensitivity but low specificity in differentiating benign from malignant IPMNs, leading to unnecessary surgeries. Discovering biomarkers is thus warranted to improve the preoperative risk stratification of IPMN. Pancreatic cyst fluid samples were obtained from patients with pathologically confirmed low-grade (n = 73) or high-grade/invasive (n = 18) IPMN. Global proteome quantitation was performed using liquid chromatography-tandem mass spectrometry. Differentially expressed proteins (DEPs) between the two groups were analyzed using Wilcoxon rank-sum test. 152 upregulated and 74 downregulated DEPs were discovered by comparing low-grade IPMN with high-grade/invasive IPMN (all p < 0.05). The enriched upstream regulators of these DEPs included let-7, miR-122, IL15, and FLT1 (p = 6.76 × 10-5 - 5.97 × 10-3). Five discriminatory biomarkers with the largest LASSO coefficients and each with AUCs of >0.75 (FAHD2A, TCEAL3, TWF1, MMUT, and NTPCR) were identified. The combined five-protein model achieved a bootstrap-corrected AUC of 0.94. A combined analysis of TCGA and GTEx databases showed TWF1 overexpression in pancreatic cancer (p = 2.22 × 10-16) that was associated with poor prognosis (p = 0.0063). The present study identified several cyst fluid proteins (particularly TWF1) that are predictive of malignant pancreatic cyst lesions. If validated in other patient populations, these biomarkers may enhance the accuracy of the preoperative detection of high-risk IPMN and thereby improve patient outcomes.
Collapse
Affiliation(s)
- Chunliang Liu
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA; Department of Gastroenterology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Amber Mosley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ehsan Irajizad
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Huangbing Wu
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Thoa Tran
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - James P Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Johannes Fahrmann
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John M DeWitt
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Samir Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - C Max Schmidt
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA.
| | - Jianjun Zhang
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA; Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA.
| |
Collapse
|
3
|
Daniel N, Farinella R, Belluomini F, Fajkic A, Rizzato C, Souček P, Campa D, Hughes DJ. The relationship of the microbiome, associated metabolites and the gut barrier with pancreatic cancer. Semin Cancer Biol 2025; 112:43-57. [PMID: 40154652 DOI: 10.1016/j.semcancer.2025.03.002] [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: 11/30/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Pancreatic cancers have high mortality and rising incidence rates which may be related to unhealthy western-type dietary and lifestyle patterns as well as increasing body weights and obesity rates. Recent data also suggest a role for the gut microbiome in the development of pancreatic cancer. Here, we review the experimental and observational evidence for the roles of the oral, gut and intratumoural microbiomes, impaired gut barrier function and exposure to inflammatory compounds as well as metabolic dysfunction as contributors to pancreatic disease with a focus on pancreatic ductal adenocarcinoma (PDAC) initiation and progression. We also highlight some emerging gut microbiome editing techniques currently being investigated in the context of pancreatic disease. Notably, while the gut microbiome is significantly altered in PDAC and its precursor diseases, its utility as a diagnostic and prognostic tool is hindered by a lack of reproducibility and the potential for reverse causality in case-control cohorts. Future research should emphasise longitudinal and mechanistic studies as well as integrating lifestyle exposure and multi-omics data to unravel complex host-microbiome interactions. This will allow for deeper aetiologic and mechanistic insights that can inform treatments and guide public health recommendations.
Collapse
Affiliation(s)
- Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | | | | | - Almir Fajkic
- Department of Pathophysiology Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | | | - Pavel Souček
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic; Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland.
| |
Collapse
|
4
|
Yoo C, Jeong H, Jeong JH, Kim KP, Lee S, Ryoo BY, Hwang DW, Lee JH, Moon DB, Kim KH, Lee SS, Song TJ, Oh D, Lee MA, Chon HJ, Lee JS, Laliotis G, Rivero-Hinojosa S, Spickard E, Renner D, Dutta P, Palsuledesai CC, Sharma S, Malhotra M, Jurdi A, Liu MC. Circulating tumor DNA status and dynamics predict recurrence in patients with resected extrahepatic cholangiocarcinoma. J Hepatol 2025; 82:861-870. [PMID: 39532185 DOI: 10.1016/j.jhep.2024.10.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/01/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND & AIMS Surgery is the only curative therapeutic option for resectable extrahepatic cholangiocarcinoma, but recurrence is common, and prognosis is poor. There is an unmet clinical need for improved decision-making regarding adjuvant chemotherapy (ACT). Herein, we evaluated the usefulness of monitoring longitudinal circulating tumor DNA (ctDNA) for molecular residual disease (MRD) in patients from the STAMP trial, which compared the efficacy of adjuvant capecitabine (CAP) vs. gemcitabine plus cisplatin (GemCis). METHODS Between July 2017 and November 2020, 101 patients were randomized 1:1 to receive GemCis (n = 50) or CAP (n = 51). Efficacy outcomes were analyzed with an extended follow-up of 19 months from the previous report. From a biomarker cohort of 89 patients, longitudinal plasma samples (n = 254) were prospectively collected post-surgery before ACT, and on-ACT at 12 and 24 weeks from cycle 1 day 1 (C1D1). ctDNA was evaluated using a personalized, tumor-informed, 16-plex PCR next-generation sequencing assay and was correlated with clinical outcomes. RESULTS In the extended follow-up analysis, median disease-free survival (DFS) and overall survival did not significantly differ between the CAP and GemCis groups. Significantly inferior DFS was associated with ctDNA positivity before ACT (hazard ratio [HR] 1.8; p = 0.029), on-ACT at 12 weeks from C1D1 (HR 7.72; p <0.001), on-ACT at 24 weeks from C1D1 (HR 5.24; p <0.001), and anytime post-surgery (HR 3.81; p <0.001). Analysis of pre-treatment to on-treatment ctDNA dynamics revealed that serially ctDNA-negative patients exhibited a significantly longer DFS compared to those with sustained ctDNA positivity (HR 6.7; p <0.001) or those who turned ctDNA positive (HR 5.8; p <0.001). CONCLUSION In patients with resected extrahepatic cholangiocarcinoma, ctDNA status and dynamics predicted recurrence during adjuvant therapy, and may help optimize clinical decision-making. IMPACT AND IMPLICATIONS The findings from this study highlight the critical role of ctDNA as a prognostic biomarker and monitoring tool for patients with resected extrahepatic cholangiocarcinoma. By demonstrating the superiority of ctDNA to predict disease recurrence compared to conventional biomarkers such as cancer antigen 19-9 and carcinoembryonic antigen, this study underscores its potential in guiding decision-making during adjuvant chemotherapy. These results may be crucial to refine post-surgical treatment strategies and improve patient outcomes. The practical application of ctDNA monitoring could lead to more personalized treatment approaches, enabling timely interventions based on molecular residual disease status. CLINICAL TRIAL REGISTRATION NUMBER NCT03079427.
Collapse
Affiliation(s)
- Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Hyehyun Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae Ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyu-Pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seonmin Lee
- University of Ulsan Digestive Disease Research Center, Seoul, Korea
| | - Baek-Yeol Ryoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dae Wook Hwang
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Hoon Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Deog-Bog Moon
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ki-Hun Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang Soo Lee
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Tae Jun Song
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dongwook Oh
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Myung Ah Lee
- Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hong Jae Chon
- Department of Medical Oncology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Ji Sung Lee
- Department of Clinical Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Xi W, Liao W, Guo T, Jiang Q, Bai X, Wu X, Feng Y, Zhang S, Wu D, Wang Q, Yang A. CA19-9/DBil: a promising indicator to distinguish between CA19-9-elevated pancreatic head-type autoimmune pancreatitis and pancreatic head cancer. BMC Gastroenterol 2025; 25:306. [PMID: 40301736 PMCID: PMC12039079 DOI: 10.1186/s12876-025-03925-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 04/22/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Pancreatic head-type autoimmune pancreatitis (PH-AIP) with elevated CA19-9 is sometimes difficult to distinguish from pancreatic head cancer (PHC) with elevated CA19-9. At times, IgG4 proves inadequate in offering assistance. The study aimed to elucidate the performance of CA19-9/DBil in distinguishing between the two conditions. METHODS This was a retrospective study. We collected serologic indicators from participants in PH-AIP and PHC Group. Three logistic regression equations were established ranging from non-adjustment (Model 1, only CA19-9/DBil included) to adjusting for sex, age, and CEA (Model 2 and Model 3) to explore the relationship between CA19-9/DBil and PH-AIP probability. ROC, Decision Curve Analysis (DCA), calibration curve were conducted. P for AUCs and net reclassification improvements (NRI) were computed to evaluate differences in discrimination and the improvement in risk reclassification between models. RESULTS The study included 90 PHC and 35 PH-AIP patients, all with elevated CA19-9. The ORs for CA19-9/DBil in three models were similar (0.915 to 0.921). ROC revealed that Model 1 had an AUC of 0.772. The sensitivity, specificity, and accuracy at the best threshold were all > 0.7. Model 1, although simple, was not inferior in its discriminative ability compared to complex models: the difference in discrimination between Model 1 and each of two adjusted models was not statistically significant (P > 0.05, both AUC and NRI). Additionally, calibration curve and DCA suggested that Model 1 had good calibration and clinical utility. CONCLUSIONS CA19-9/DBil exhibited promising diagnostic performance in differentiating between CA19-9-elevated PH-AIP and PHC.
Collapse
Affiliation(s)
- Wenfeng Xi
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Wanying Liao
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Tao Guo
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.
| | - Qingwei Jiang
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyin Bai
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xi Wu
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yunlu Feng
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Shengyu Zhang
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Dongsheng Wu
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Qiang Wang
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Aiming Yang
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Ivaskiene T, Kaspute G, Ramanavicius A, Prentice U. Molecularly Imprinted Polymer Advanced Hydrogels as Tools for Gastrointestinal Diagnostics. Gels 2025; 11:269. [PMID: 40277704 PMCID: PMC12026608 DOI: 10.3390/gels11040269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Gastroenterology faces significant challenges due to the global burden of gastrointestinal (GI) diseases, driven by socio-economic disparities and their wide-ranging impact on health and healthcare systems. Advances in molecularly imprinted polymers (MIPs) offer promising opportunities for developing non-invasive, cost-effective diagnostic tools that enhance the accuracy and accessibility of GI disease detection. This research explores the potential of MIP-based sensors in revolutionizing gastrointestinal diagnostics and improving early detection and disease management. Biomarkers are vital in diagnosing, monitoring, and personalizing disease treatment, particularly in gastroenterology, where advancements like MIPs offer highly selective and non-invasive diagnostic solutions. MIPs mimic natural recognition mechanisms, providing stability and sensitivity even in complex biological environments, making them ideal for early disease detection and real-time monitoring. Their integration with advanced technologies, including conducting polymers, enhances their functionality, enabling rapid, point-of-care diagnostics for gastrointestinal disorders. Despite regulatory approval and scalability challenges, ongoing innovations promise to revolutionize diagnostics and improve patient outcomes through precise approaches.
Collapse
Affiliation(s)
- Tatjana Ivaskiene
- State Research Institute Centre for Innovative Medicine, LT-08410 Vilnius, Lithuania; (T.I.); (G.K.)
| | - Greta Kaspute
- State Research Institute Centre for Innovative Medicine, LT-08410 Vilnius, Lithuania; (T.I.); (G.K.)
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), LT-10257 Vilnius, Lithuania
| | - Arunas Ramanavicius
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), LT-10257 Vilnius, Lithuania
- Department of Physical Chemistry, Faculty of Chemistry and Geosciences, Institute of Chemistry, Vilnius University, LT-03225 Vilnius, Lithuania
| | - Urte Prentice
- State Research Institute Centre for Innovative Medicine, LT-08410 Vilnius, Lithuania; (T.I.); (G.K.)
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), LT-10257 Vilnius, Lithuania
- Department of Physical Chemistry, Faculty of Chemistry and Geosciences, Institute of Chemistry, Vilnius University, LT-03225 Vilnius, Lithuania
| |
Collapse
|
7
|
Chun JW, Lee DE, Han N, Heo S, Kim H, Lee MR, Park HM, Han SS, Park SJ, Kim TH, Lee WJ, Kim YH, Kong SY, Woo SM. Mutant KRAS and GATA6 Stratify Survival in Patients Treated with Chemotherapy for Pancreatic Adenocarcinoma: A Prospective Cohort Study. Cancers (Basel) 2025; 17:896. [PMID: 40075743 PMCID: PMC11899085 DOI: 10.3390/cancers17050896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Several pancreatic adenocarcinoma (PA) biomarkers beyond the traditional carbohydrate antigen (CA)19-9 have been identified but are lacking large-scale prospective validation. This prospective cohort study evaluated the prognostic impact of potential PA biomarkers. METHODS We enrolled 238 of 288 patients with histologically proven PA. We assessed candidate biomarkers, including CA19-9, germline BRCA1/2, and ATM mutations, as well as mutant KRAS circulating tumor DNA (ctDNA) in blood samples. Additionally, we evaluated the expression of SLC29A1 (hENT1), DCK, CES2, and GATA6. We examined the association of candidate biomarkers with progression-free survival (PFS) and overall survival (OS). RESULTS We analyzed biomarker efficacy in 200 (median age 65 years; 55% male) of the enrolled patients who received chemotherapy. A high mutant KRAS ctDNA concentration (hazard ratio [HR]: 1.508 and 95% confidence interval [CI]: 1.052-2.161 for PFS; HR: 1.796 and 95% CI: 1.203-2.681 for OS) and high CA19-9 level (HR: 1.647 and 95% CI: 1.177-2.306 for PFS; HR: 1.803 and 95% CI: 1.248-2.605 for OS) were associated with poor prognosis. High GATA6 RNA expression was linked to longer PFS (HR: 0.336 and 95% CI: 0.195-0.582) and OS (HR: 0.304 and 95% CI: 0.165-0.560). CONCLUSIONS Plasma mutant KRAS ctDNA concentrations and GATA6 expression could serve as significant prognostic biomarkers in patients with PA, potentially guiding therapeutic decisions and prognostication.
Collapse
Affiliation(s)
- Jung Won Chun
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Cancer Biomedical Science, National Cancer Center, Graduate School of Cancer Science and Policy, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Dong-eun Lee
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Nayoung Han
- Department of Pathology, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - SooBeen Heo
- Targeted Therapy Branch, Center for Rare Cancers, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Hyeji Kim
- Targeted Therapy Branch, Center for Rare Cancers, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Mi Rim Lee
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Hyeong Min Park
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Sung-Sik Han
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Cancer Biomedical Science, National Cancer Center, Graduate School of Cancer Science and Policy, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Sang-Jae Park
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Tae Hyun Kim
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Woo Jin Lee
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Yun-Hee Kim
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Cancer Biomedical Science, National Cancer Center, Graduate School of Cancer Science and Policy, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Sun-Young Kong
- Cancer Biomedical Science, National Cancer Center, Graduate School of Cancer Science and Policy, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Targeted Therapy Branch, Center for Rare Cancers, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Department of Laboratory Medicine, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Sang Myung Woo
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Cancer Biomedical Science, National Cancer Center, Graduate School of Cancer Science and Policy, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| |
Collapse
|
8
|
Heller M, Mann DA, Katona BW. Current Approaches of Pancreatic Cancer Surveillance in High-Risk Individuals. J Gastrointest Cancer 2025; 56:61. [PMID: 39932614 PMCID: PMC11814005 DOI: 10.1007/s12029-025-01184-1] [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] [Accepted: 01/25/2025] [Indexed: 02/14/2025]
Abstract
Currently, those recommended to undergo pancreatic cancer (PC) surveillance include appropriately aged individuals at high risk of PC due to an identifiable genetic susceptibility or those without identifiable genetic susceptibility who nonetheless have a strong family history of PC. With increases in identification of individuals at high risk for PC and increased use of PC surveillance in clinical practice, there has been increasing debate about who should undergo surveillance as well as how surveillance should be performed including use of imaging and blood-based testing. Furthermore, there is increasing interest in the outcomes of PC surveillance in high-risk individuals with some studies demonstrating that surveillance leads to downstaging of PC and improvements in survival. In this review, we summarize the current state of PC surveillance in high-risk individuals, providing an overview of the risk factors associated with PC, selection of high-risk individuals for PC surveillance, and the current, but non-uniform, recommendations for performing PC surveillance. Additionally, we review approaches to apply various imaging and blood-based tests to surveillance and the outcomes of PC surveillance.
Collapse
Affiliation(s)
- Melissa Heller
- Division of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Derek A Mann
- Division of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bryson W Katona
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd., 751 South Pavilion, Philadelphia, PA, 19104, USA.
| |
Collapse
|
9
|
Kwon YJ, Min JH, Hwang JA, Kim SH, Kim YK, Kim H, Gu K, Lee JH, Shin J, Choi SY, Baek SY. Clinical significance of CA 19-9 elevation during postoperative surveillance for extrahepatic bile duct cancer: a nomogram-based approach for the prediction of short-term recurrence. HPB (Oxford) 2025; 27:195-205. [PMID: 39586759 DOI: 10.1016/j.hpb.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/13/2024] [Accepted: 10/29/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND This study aimed to assess the significance of elevated carbohydrate antigen (CA) 19-9 in postoperative surveillance of extrahepatic bile duct cancer and to identify short-term recurrence predictors. METHODS This retrospective study included patients with elevated CA 19-9 post-curative surgery. Patients were categorized into positive and negative CT groups based on the detection of recurrence at CA 19-9 elevation. Short-term recurrence was defined as recurrence within 6 months in the negative CT group. We identified the factors associated with short-term recurrence and devised a predictive nomogram. RESULTS Among the 190 patients, 91 (47.9 %) exhibited tumor recurrence with CA 19-9 elevation (CT-positive group), whereas 99 (52.1 %) showed no recurrence (CT-negative group). In the CT-negative group (n = 99), 22 (22.2 %) experienced short-term tumor recurrence within 6 months. Preoperative CA 19-9 (odds ratio [OR]: 1.5, p = 0.016), postoperative CA 19-9 (OR: 1.9, p = 0.047), adjuvant treatment (OR: 3.5, p = 0.032), and the absence of inflammation (OR: 3.5, p = 0.045) were predictors of short-term recurrence. The area under the curve of the nomogram was 0.80 (95 % CI: 0.69-0.90). CONCLUSION Despite elevated CA 19-9 levels, approximately 50 % of patients exhibited no recurrence during postoperative surveillance for extrahepatic bile duct cancer. Factors influencing short-term recurrence encompass pre- and postoperative CA 19-9, adjuvant treatment, and inflammatory status.
Collapse
Affiliation(s)
- Yong Jae Kwon
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ji Hye Min
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Jeong Ah Hwang
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seong Hyun Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young Kon Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Honsoul Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyowon Gu
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeong Hyun Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jaeseung Shin
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seo-Youn Choi
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sun-Young Baek
- Biomedical Statistics Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| |
Collapse
|
10
|
Dumitrascu T. An Isolated Hydatid Cyst of the Spleen with High Serum Levels of CA 19-9-A Meaningful Association or Just a Challenge for Diagnosis? A Case Report. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:182. [PMID: 40005300 PMCID: PMC11857084 DOI: 10.3390/medicina61020182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025]
Abstract
An isolated hydatid cyst of the spleen represents an exceptional pathology, and its association with high CA 19-9 serum levels was not previously reported. This case presents a patient with an isolated hydatid cyst of the spleen, with preoperative high CA 19-9 serum levels in the absence of other pathologies and normalization of CA 19-9 serum levels after surgery (i.e., splenectomy). The source and clinical value of high serum levels of CA 19-9 in hydatid cysts of the spleen remains unclear. High serum levels of CA 19-9 in the context of a splenic cyst may complicate diagnosis and challenge the therapeutic strategy.
Collapse
Affiliation(s)
- Traian Dumitrascu
- Department of General Surgery, Division of Surgical Oncology, Fundeni Clinical Institute, Carol Davila University of Medicine and Pharmacy, Fundeni Street No 258, 022328 Bucharest, Romania
| |
Collapse
|
11
|
Götze J, Meißner K, Pereira-Veiga T, Belloum Y, Schneegans S, Kropidlowski J, Gorgulho J, Busch A, Honselmann KC, Schönrock M, Putscher A, Peine S, Nitschke C, Simon R, Spindler V, Izbicki JR, Hackert T, Bokemeyer C, Pantel K, Uzunoglu FG, Sinn M, Wikman H. Identification and characterization of tumor and stromal derived liquid biopsy analytes in pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res 2025; 44:14. [PMID: 39815324 PMCID: PMC11737273 DOI: 10.1186/s13046-024-03262-x] [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: 09/05/2024] [Accepted: 12/19/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND The lack of predictive biomarkers contributes notably to the poor outcomes of patients with pancreatic ductal adenocarcinoma (PDAC). Cancer-associated fibroblasts (CAFs) are the key components of the prominent PDAC stroma. Data on clinical relevance of CAFs entering the bloodstream, known as circulating CAFs (cCAFs) are scarce. Here, we developed a combined liquid biopsy assay to detect cCAFs and circulating tumor cells (CTCs) in metastatic PDAC (mPDAC) and other metastatic gastrointestinal malignancies (mGI). In addition, we evaluated plasma hyaluronan (HA) levels as a complementary surrogate biomarker of the stromal extent in patients with PDAC. METHODS A sequential liquid biopsy assay based on a two step-enrichment, combining marker dependent and independent cell enrichment, was established for cCAF and CTC detection and validated in mPDAC and mGI patients. The enriched cells were identified by multiplex immunofluorescence. HA measurement was performed by ELISA on blood samples from healthy blood donors (HD), localized and late-stage PDAC patients. RESULTS cCAFs (≥ 1cCAFs/7.5 mL blood) were detected in 95.4% of mPDAC and in 78.2% of mGI patients, with significantly higher numbers in mPDAC compared to mGI patients (mean number 22.7 vs. 11.0; P = 0.0318). mPDAC patients with ≥ 15 cCAFs/7.5 mL blood had a significant shorter median overall survival (mOS 3.2 months (95% confidence interval (CI) 0.801-5.855) vs. 14.2 months (95% CI 6.055-22.332); P = 0.013), whereby CTC levels were not associated with mOS. In mGI neither cCAFs nor CTCs had a significant impact on OS. HA plasma levels in mPDAC patients were significantly higher compared to HD (mean 123.0 ng/mL vs. 74.45 ng/mL, P = 0.015). High HA in localized and late-stage PDAC were associated with a significantly shorter mOS (mOSlocalized PDAC: 12.6 months vs. 23.5 months (P = 0.008); mOSmPDAC: 1.8 months vs. 5.3 months (P = 0.004)). CONCLUSIONS Our liquid biopsy assay provides robust detection of cCAFs in mPDAC and mGI patients. The measurement of both circulatory stromal parameters, cCAFs and HA, adds valuable clinical information as they are associated with an unfavorable outcome in PDAC. These results highlight that stromal characteristics unique to PDAC could be leveraged to fill the current gap in discovering predictive biomarkers.
Collapse
Affiliation(s)
- Julian Götze
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany.
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany.
| | - Kira Meißner
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Thais Pereira-Veiga
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Yassine Belloum
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Svenja Schneegans
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Jolanthe Kropidlowski
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Joao Gorgulho
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany
| | - Alina Busch
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany
| | - Kim Christin Honselmann
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Martin Schönrock
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany
| | - Arne Putscher
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Sven Peine
- Department of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Nitschke
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volker Spindler
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob Robert Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Faik Güntaç Uzunoglu
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marianne Sinn
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany.
| | - Harriet Wikman
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany.
| |
Collapse
|
12
|
Dhingra S, Raman P, Ramsaroop T, Harrison I, Bergsten T, Nusbaum E, Feldman LE. Elevated serum CA 19-9 level mimicking pancreaticobiliary carcinoma from a hepatic abscess: case report and literature review. Front Med (Lausanne) 2025; 11:1470046. [PMID: 39876872 PMCID: PMC11772410 DOI: 10.3389/fmed.2024.1470046] [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: 07/24/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025] Open
Abstract
Serum levels of the tumor marker CA 19-9 are widely utilized in the diagnosis and monitoring pancreatic and biliary malignancies. However, serum levels of CA 19-9 have also been reportedly elevated in non-malignant conditions. Here, we present the rare case of a 65-year-old woman with a history of gallbladder malignancy who was found to have a new hepatic lesion on surveillance CT with an associated elevation in CA 19-9 to 5,866 U/mL. Drainage of the lesion and treatment with antibiotics resulted in a rapid decline in CA 19-9 levels, indicating that the elevation in CA 19-9 was due to a benign hepatic lesion. We review eight similar reported cases of CA 19-9 elevations due to benign hepatic abscesses, thereby highlighting a need to interpret the tumor marker with caution.
Collapse
Affiliation(s)
- Shaurya Dhingra
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Puneet Raman
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Taylor Ramsaroop
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Isaiah Harrison
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Tova Bergsten
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Erin Nusbaum
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Lawrence E. Feldman
- University of Illinois College of Medicine, Chicago, IL, United States
- Jesse Brown VA Medical Center, Chicago, IL, United States
| |
Collapse
|
13
|
Lopes M, Figueiredo V, Mendes A, Amaral M, Delgado Alves J. Lung Adenocarcinoma Presenting With Elevated Serum Carbohydrate Antigen 19-9 Levels: A Case Report. Cureus 2025; 17:e77786. [PMID: 39981495 PMCID: PMC11841819 DOI: 10.7759/cureus.77786] [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] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
Carbohydrate antigen 19-9 (CA 19-9) is a tumor marker usually used for the diagnosis and follow-up of pancreatic, gastric, and hepatobiliary malignancies. However, it has a low specificity and can be elevated in a wide array of other conditions. CA 19-9 elevation in lung tumors seems to be associated with the worst prognosis, but its role in this condition is not fully established yet. We present the case of a 78-year-old woman with mild consumptive symptoms, consisting of unselective anorexia and weight loss, and isolated high levels of CA 19-9. Hepatobiliary and gastrointestinal lesions were excluded, and a suspicious lesion was found in the right lung. A biopsy, which was only possible several months after presentation due to the location and small dimensions of the lesion, confirmed the diagnosis of lung adenocarcinoma. After an initial indolent course, the patient evolved with disease progression and is currently under treatment with palliative chemotherapy. This case illustrates the thorough investigation performed to clarify the elevation of a tumor biomarker measured in a pauci-symptomatic patient, which led to the identification of a rare and unsuspected etiology of high levels of CA 19-9.
Collapse
Affiliation(s)
- Mónica Lopes
- Department of Internal Medicine IV, Hospital Professor Doutor Fernando Fonseca, Amadora, PRT
| | - Vera Figueiredo
- Department of Pulmonology, Hospital Professor Doutor Fernando Fonseca, Amadora, PRT
| | - Ana Mendes
- Department of Oncology, Hospital Professor Doutor Fernando Fonseca, Amadora, PRT
| | - Marta Amaral
- Department of Internal Medicine IV, Hospital Professor Doutor Fernando Fonseca, Amadora, PRT
| | - José Delgado Alves
- Department of Internal Medicine IV, Hospital Professor Doutor Fernando Fonseca, Amadora, PRT
| |
Collapse
|
14
|
Yu J, He AR, Ouf M, Mehta R, Anaya DA, Denbo J, Bridges C, Tin A, Aushev VN, Palsuledesai CC, Sharma S, Jurdi A, Liu MC, Kim RD. Detecting Early Recurrence With Circulating Tumor DNA in Stage I-III Biliary Tract Cancer After Curative Resection. JCO Precis Oncol 2025; 9:e2400443. [PMID: 39772829 PMCID: PMC11723488 DOI: 10.1200/po-24-00443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/09/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
PURPOSE This study aimed to assess (1) the prognostic value of circulating tumor DNA (ctDNA) and (2) the ability of ctDNA to detect recurrence compared with standard surveillance in curatively resected early-stage biliary tract cancer (BTC). METHODS This retrospective, multicenter cohort study evaluated serial ctDNA testing for surveillance in patients with early-stage BTC after curative resection. We evaluated the relapse-free survival (RFS) by ctDNA positivity. The sensitivity of ctDNA in detecting a confirmed recurrence of BTC, defined as a biopsy-proven or true progression by radiographic tumor dynamics, was evaluated. The lead time was calculated from the first ctDNA detection to the confirmed recurrence. RESULTS A total of 56 patients with curatively resected stage I-III BTC were included in this study, with a median follow-up of 12.8 months from the date of surgery. ctDNA detection during the molecular residual disease window period (median RFS, 6.6 months v not reached; hazard ratio [HR], 26 [95% CI, 2.6 to 265]; P < .0001) and during the surveillance period (median RFS, 19.3 months v not reached; HR, 20 [95% CI, 2.6 to 153]; P < .0001) were associated with poorer RFS. Sixteen patients had confirmed recurrence. ctDNA identified recurrence in 93.8.% (15/16) of the recurred patients with an average lead time of 3.7 months. Carbohydrate antigen 19-9 levels did not show any significant correlation with RFS (HR, 1.17 [95% Cl, 0.24 to 5.71]; P = .844) in contrast to ctDNA. CONCLUSION The findings from our real-world cohort study revealed the (1) promising value of ctDNA as a prognostic biomarker for relapse in curatively resected BTC and (2) potential early detectability of recurrence by ctDNA compared with standard surveillance.
Collapse
Affiliation(s)
- James Yu
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Aiwu Ruth He
- Department of Gastrointestinal Oncology, MedStar Georgetown University Hospital, Washington, DC
| | - Mahmoud Ouf
- Department of Gastrointestinal Oncology, MedStar Georgetown University Hospital, Washington, DC
| | - Rutika Mehta
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Daniel A. Anaya
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Jason Denbo
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL
| | | | | | | | | | | | | | | | - Richard D. Kim
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
15
|
Yu L, Wang H, Wang F, Guo J, Xiao B, Hou Z, Lu Z, Pan Z, Zhou Y, Ye S, Wan D, Lin B, Ou Q, Fang Y. Serum biomarkers REG1A and REG3A combined with the traditional CEA represent a novel nomogram for the screening and risk stratification of colorectal cancer. Clin Transl Oncol 2025; 27:277-290. [PMID: 38965192 DOI: 10.1007/s12094-024-03566-6] [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: 05/04/2024] [Accepted: 06/09/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND To develop and validate a serum protein nomogram for colorectal cancer (CRC) screening. METHODS The serum protein characteristics were extracted from an independent sample containing 30 colorectal cancer and 12 polyp tissues along with their paired samples, and different serum protein expression profiles were validated using RNA microarrays. The prediction model was developed in a training cohort that included 1345 patients clinicopathologically confirmed CRC and 518 normal participants, and data were gathered from November 2011 to January 2017. The lasso logistic regression model was employed for features selection and serum nomogram building. An internal validation cohort containing 576 CRC patients and 222 normal participants was assessed. RESULTS Serum signatures containing 27 secreted proteins were significantly differentially expressed in polyps and CRC compared to paired normal tissue, and REG family proteins were selected as potential predictors. The C-index of the nomogram1 (based on Lasso logistic regression model) which contains REG1A, REG3A, CEA and age was 0.913 (95% CI, 0.899 to 0.928) and was well calibrated. Addition of CA199 to the nomogram failed to show incremental prognostic value, as shown in nomogram2 (based on logistic regression model). Application of the nomogram1 in the independent validation cohort had similar discrimination (C-index, 0.912 [95% CI, 0.890 to 0.934]) and good calibration. The decision curve (DCA) and clinical impact curve (ICI) analysis demonstrated that nomogram1 was clinically useful. CONCLUSIONS This study presents a serum nomogram that included REG1A, REG3A, CEA and age, which can be convenient for screening of colorectal cancer.
Collapse
Affiliation(s)
- Long Yu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Hao Wang
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Fulong Wang
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Jian Guo
- Senboll Biotechnology Co., Ltd., Pingshan Bio-Pharmacy Business Accelerator, Pingshan District, Shenzhen, 518000, Guangdong, China
| | - Binyi Xiao
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Zhenlin Hou
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Zhenhai Lu
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Zhizhong Pan
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Yaxian Zhou
- Senboll Biotechnology Co., Ltd., Pingshan Bio-Pharmacy Business Accelerator, Pingshan District, Shenzhen, 518000, Guangdong, China
| | - Sibin Ye
- Senboll Biotechnology Co., Ltd., Pingshan Bio-Pharmacy Business Accelerator, Pingshan District, Shenzhen, 518000, Guangdong, China
| | - Desen Wan
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Bo Lin
- Department of Thyroid Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Qingjian Ou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
| | - Yujing Fang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
| |
Collapse
|
16
|
Jin Y, Hu R, Gu Y, Wei A, Li A, Zhang Y. Quantitative site-specific N-glycosylation analysis reveals IgG glyco-signatures for pancreatic cancer diagnosis. Clin Proteomics 2024; 21:68. [PMID: 39734184 DOI: 10.1186/s12014-024-09522-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024] Open
Abstract
BACKGROUND Pancreatic cancer is a highly aggressive tumor with a poor prognosis due to a low early detection rate and a lack of biomarkers. Most of pancreatic cancer is pancreatic ductal adenocarcinoma (PDAC). Alterations in the N-glycosylation of plasma immunoglobulin G (IgG) have been shown to be closely associated with the onset and development of several cancers and could be used as biomarkers for diagnosis. The study aimed to explore intact N-glycosylation profile of IgG in patients with PDAC and find relation between intact N-glycosylation profile of IgG and clinical information such as diagnosis and prognosis. METHODS In this study, we employed a well-evaluated approach (termed GlycoQuant) to assess the site-specific N-glycosylation profile of human plasma IgG in both healthy individuals and patients with pancreatic ductal adenocarcinoma (PDAC). The datasets generated and analyzed during the current study are available in the ProteomeXchange Consortium ( http://www.proteomexchange.org/ ) via the iProX partner repository, with the dataset identifier PXD051436. RESULTS The analysis of rapidly purified IgG samples from 100 patients with different stages of PDAC, in addition to 30 healthy controls, revealed that the combination of carbohydrate antigen 19 - 9 (CA19-9), IgG1-GP05 (IgG1-TKPREEQYNSTYR-HexNAc [4]Hex [5]Fuc [1]NeuAc [1]), and IgG4-GP04 (IgG4-EEQFNSTYR- HexNAc [4]Hex [5]Fuc [1]NeuAc [1]) can be used to distinguish between PDAC patients and healthy individuals (AUC = 0.988). In addition, cross validation of the diagnosis model showed satisfactory result. CONCLUSIONS The study demonstrated that the integrated quantitative method can be utilized for large-scale clinical N-glycosylation research to identify novel N-glycosylated biomarkers. This could facilitate the development of clinical glycoproteomics.
Collapse
Affiliation(s)
- Yi Jin
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan, 610041, China
| | - Ran Hu
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan, 610041, China
| | - Yufan Gu
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, Keyuan 4th Road, Gaopeng Avenue, Hi-tech Zone, Chengdu, Sichuan, 610041, China
| | - Ailin Wei
- Guang'an People's Hospital, Guang'an, 638001, China
| | - Ang Li
- Guang'an People's Hospital, Guang'an, 638001, China.
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan, 610041, China.
| | - Yong Zhang
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, Keyuan 4th Road, Gaopeng Avenue, Hi-tech Zone, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
17
|
Kim HS, Kim W, Yun WG, Jung HS, Han Y, Lee M, Kwon W, Jang JY, Park JS. Novel Predictive Strategy Using CA19-9 and Fecal Elastase Levels to Make Treatment Decisions for Resectable Pancreatic Cancer: A Retrospective Study. Biomedicines 2024; 13:62. [PMID: 39857647 PMCID: PMC11763034 DOI: 10.3390/biomedicines13010062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Carbohydrate antigen 19-9 (CA19-9) is used as a marker to predict recurrence and survival of patients with pancreatic ductal adenocarcinoma (PDAC). Recently, fecal elastase-1 (FE-1) has been shown to correlate with prognosis in patients with PDAC. Method: A total of 536 patients who underwent curative intent surgery between 2010 and 2019 were included in the study. The cutoff points of preoperative CA19-9 and FE-1 levels were extracted from the Youden index and previous studies. Cox proportional hazard models were used to investigate the association between preoperative tumor marker levels and survival after surgery. Results: Patients with CA19-9 ≥ 385 had more advanced T-/N-stages and lower survival rates compared to those with CA19-9 < 385. Multivariate Cox analyses demonstrated that combining preoperative tumor markers was associated with worse 3-year overall survival (both CA19-9 and FE-1 low, HR = 1.41, p = 0.044; both high, HR = 1.44, p = 0.047; CA19-9 high and FE-1 low, HR = 2.00, p < 0.001; and p for trend < 0.001). The same trend was confirmed in the analysis with recurrence-free survival. Conclusions: This study presents a new predictive strategy using combined CA19-9 and FE-1 levels to determine the treatment for resectable pancreatic cancer.
Collapse
Affiliation(s)
- Hyung Sun Kim
- Pancreatobiliary Cancer Clinic, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Woojin Kim
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Korea Medical Institute, Seoul 04522, Republic of Korea
| | - Won-Gun Yun
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hye-Sol Jung
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Youngmin Han
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Mirang Lee
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Joon Seong Park
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
18
|
Khasawneh H, O'Brien C, Czeyda-Pommersheim F, Qayyum A, Miller FH, Arif Tiwari H, Paspulati RM, Kierans AS. Beyond cholangiocarcinoma: imaging features of mimicking pathologies in the biliary tract. Abdom Radiol (NY) 2024:10.1007/s00261-024-04749-z. [PMID: 39710762 DOI: 10.1007/s00261-024-04749-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: 10/16/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/24/2024]
Abstract
Cholangiocarcinoma (CCA) is the second most common primary malignancy of the hepatobiliary system and presents as a heterogeneous disease with three distinct morphological subtypes: mass-forming, periductal-infiltrating, and intraductal-growing, each characterized by distinguishing imaging features. Accurate diagnosis of CCA is challenging due to the overlap of imaging findings with a broad range of benign and malignant conditions. Therefore, it is essential for radiologists to recognize these mimickers and offer a reasonable differential diagnosis, as this has a significant impact on patient management. Although histopathological confirmation is often required for a definitive diagnosis, understanding specific imaging characteristics that differentiate CCA from its mimickers is crucial. This article highlights a variety of benign and malignant conditions that resemble CCA on imaging, emphasizing features that enhance diagnostic accuracy in clinical practice.
Collapse
Affiliation(s)
- Hala Khasawneh
- The University of Texas Southwestern Medical Center, Dallas, USA.
| | | | | | | | - Frank H Miller
- Northwestern University Feinberg School of Medicine, Chicago, USA
| | | | | | | |
Collapse
|
19
|
Gao W, Huang YS, Wang YD. Construction and validation of a predictive model for the risk of rebleeding in patients with esophageal and gastric varices hemorrhage. BMC Gastroenterol 2024; 24:471. [PMID: 39716072 DOI: 10.1186/s12876-024-03569-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND AND PURPOSE Esophageal and gastric varices hemorrhage (EGVH) is a life-threatening condition with the 6-week mortality rate of 15-25%. Up to 60% of patients with EGVH may experience rebleeding with a mortality rate of 33%. The existing scoring systems, such as RS scoring system (Rockall score, RS) and GBS scoring system (Glasgow-Blatchford score, GBS), have limitations in predicting the risk of rebleeding. Our study was to construct and validate a novel predictive model for the risk of rebleeding in patients with EGVH and to compare the predictive power of the predictive model with GBS and pRS. METHODS Data of patients with EGVH was collected in the First Affiliated Hospital of Dalian Medical University from January 2016 to June 2020. Binary logistic and stepwise regression was performed to construct a predictive model. We compared the predictive power of the new predictive model to the GBS and pRS scoring systems. RESULTS Clinical data from a total of 265 patients with EGVH was collected. Six factors including systolic blood pressure, transfusion requirement, CA199, platelet count, upper esophageal varices and severity of esophageal varices were included in our new predictive model. The AUCs of the specificity of the predictive model, GBS and pRS are 0.82, 0.60 and 0.56. CONCLUSION This study successfully constructed a predictive model for the risk of rebleeding in patients with EGVH. This predictive model demonstrated higher predictive ability than pRS and GBS scoring systems for assessing rebleeding risk in EGVH patients.
Collapse
Affiliation(s)
- Wei Gao
- Department of Gastroenterology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China
| | - Yu-Shuang Huang
- Dalian Public Health Clinical Center, Liaoning Province, Dalian, China
| | - Ying-De Wang
- Department of Gastroenterology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China.
| |
Collapse
|
20
|
Tanaka H, Mise Y, Takahashi A, Kawano F, Takeda Y, Imamura H, Ichida H, Yoshioka R, Saiura A. Analyzing the high frequency of false-positive carcinoembryonic antigen elevations in postoperative pancreatic ductal adenocarcinoma. Langenbecks Arch Surg 2024; 410:6. [PMID: 39672933 DOI: 10.1007/s00423-024-03577-5] [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: 10/15/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
PURPOSE The dynamics of postoperative carcinoembryonic antigen (CEA) in pancreatic ductal adenocarcinoma (PDAC) patients have not been well assessed. This study investigated the correlation between postoperative CEA elevations and tumor recurrence. METHODS Medical records were retrospectively analyzed for 84 patients who received curative resection for PDAC from January 2019 to December 2020. Postoperative CEA levels were monitored for a minimum of 12 months. False-positive CEA elevation was defined as a CEA level exceeding 5 ng/mL without evidence of recurrence in imaging studies. RESULTS Of the examined patients, 59 (70%) exhibited CEA > 5 ng/mL within the observation period. The sensitivity and specificity of elevated CEA levels for detecting recurrence were 84% and 41%, respectively. CEA elevations without tumor recurrence were observed in 27 patients, indicating a false-positive rate of 59%. More than half of these patients demonstrated peak CEA levels between 5 and 10 ng/mL, while only true-positive patients exhibited CEA levels exceeding 40.0 ng/mL. CONCLUSION CEA may rise in more than half of postoperative PDAC patients without recurrence. CEA alone is not a robust postoperative marker.
Collapse
Affiliation(s)
- Haruka Tanaka
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yoshihiro Mise
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Atsushi Takahashi
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Fumihiro Kawano
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yoshinori Takeda
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hiroshi Imamura
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hirofumi Ichida
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Ryuji Yoshioka
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Akio Saiura
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
21
|
Saha B, Chakravarty S, Ray S, Saha H, Das K, Ghosh I, Mallick B, Biswas NK, Goswami S. Correlating tissue and plasma‑specific piRNA changes to predict their possible role in pancreatic malignancy and chronic inflammation. Biomed Rep 2024; 21:186. [PMID: 39420923 PMCID: PMC11484194 DOI: 10.3892/br.2024.1874] [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: 07/01/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
The aggressiveness of pancreatic ductal adenocarcinoma is primarily due to lack of effective early detection biomarkers. Circulating non-coding RNAs serve as diagnostic or prognostic biomarkers in multiple types of cancer. Comparison of their expression between diseased tissue and relevant body fluids such as saliva, urine, bile, pancreatic juice, blood etc. may reveal mechanistic involvement of common non-coding RNAs. piwi-interacting RNAs (piRNAs) are a class of non-coding RNAs. The aim of the present study was to investigate plasma and tumour tissue piRNA changes in patients with pancreatic cancer (PC) and explore the possible role in tumorigenesis and pancreatic inflammation. Sequencing of circulating plasma small RNAs from patients with PC and chronic pancreatitis (CP) was performed and differentially expressed piRNAs were compared with those in tissues. Subsequent search for target genes for those piRNAs was performed followed by pathway and cluster analysis. A total of 36 piRNAs were shown to be deregulated in pancreatic tumour tissue and alteration of 11 piRNAs was detected in plasma of patients with PC. piRNAs hsa-piR-23246, hsa-piR-32858 and hsa-piR-9137 may serve a key role in PC development as their expression was correlated in both plasma and tumour tissue. Key piRNA-target interactions interfering with key biological pathways were also characterized. A total of 19 deregulated piRNAs in plasma samples of patients with CP was identified; these targeted genes responsible for chronic inflammation. Therefore, the present study provides a comprehensive description of piRNA alteration in pancreatic malignancy and inflammation; these may be explored for biomarker potential in future.
Collapse
Affiliation(s)
- Barsha Saha
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, Nadia, West Bengal 741251, India
- Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad 121001, India
| | - Shouvik Chakravarty
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, Nadia, West Bengal 741251, India
- Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad 121001, India
| | - Sukanta Ray
- Institute of Postgraduate Medical Education & Research, Kolkata 700020, India
| | - Hemabha Saha
- Institute of Postgraduate Medical Education & Research, Kolkata 700020, India
| | - Kshaunish Das
- Institute of Postgraduate Medical Education & Research, Kolkata 700020, India
| | - Indranil Ghosh
- Chittaranjan National Cancer Institute, Kolkata 700026, India
| | | | - Nidhan K. Biswas
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, Nadia, West Bengal 741251, India
| | - Srikanta Goswami
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, Nadia, West Bengal 741251, India
| |
Collapse
|
22
|
Chang J, Zhang L, Li Z, Qian C, Du J. Exosomal non-coding RNAs (ncRNAs) as potential biomarkers in tumor early diagnosis. Biochim Biophys Acta Rev Cancer 2024; 1879:189188. [PMID: 39313040 DOI: 10.1016/j.bbcan.2024.189188] [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: 06/13/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 09/25/2024]
Abstract
Exosomes, extracellular vesicles carrying a cargo rich in various non-coding RNAs (ncRNAs), have emerged as crucial mediators of intercellular communication. Their stability, abundance, and specificity make exosomal ncRNAs promising candidates for biomarker discovery. The discovery of exosomal ncRNAs has unveiled a novel avenue for the exploration of biomarkers in tumor early diagnosis. This review consolidates current knowledge on the role of exosomal ncRNAs as potential biomarkers in the early detection of various tumors. We provide an overview of recent studies demonstrating the diagnostic potential of exosomal ncRNAs across multiple cancer types, highlighting their sensitivity, specificity, and feasibility for early detection. This review underscores the potential of exosomal ncRNAs as non-invasive biomarkers for early tumor diagnosis, paving the way for improved clinical outcomes through timely intervention and personalized management strategies.
Collapse
Affiliation(s)
- Jingyue Chang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong, China
| | - Lingquan Zhang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong, China
| | - Zeting Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong, China
| | - Chungen Qian
- Department of Reagent Research and Development, Shenzhen YHLO Biotech Co., Ltd., Shenzhen 518172, Guangdong, China.
| | - Juan Du
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong, China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong, China.
| |
Collapse
|
23
|
Pillai S, Antony T, Godla UR, Bhaskar E. Infection of the ovarian cyst: a rare presentation of extraintestinal salmonellosis. BMJ Case Rep 2024; 17:e261371. [PMID: 39306340 DOI: 10.1136/bcr-2024-261371] [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: 01/26/2025] Open
Abstract
A woman in her early 20's presented with fever and unintentional weight loss of 4 kg over a period of 1 month and abdominal pain for 10 days. Empirical antibiotic therapy administered prior to hospitalisation was not successful. Evaluation for fever was unrewarding except for an abnormal ultrasound which showed two cysts with the largest dimension of 9 cm in the right adnexal region. All blood cultures were sterile. She underwent laparoscopic cystectomy. Bacterial culture of cyst fluid grew Salmonella enterica subspecies enterica serotype Typhi which was found to be resistant to fluoroquinolones. The case emphasises the fact that localised infection of the ovarian cyst can occur in extraintestinal salmonellosis that can have a negative blood culture and can mimic ovarian malignancy.
Collapse
Affiliation(s)
- Sharanya Pillai
- Microbiology, Sri Ramachandra Medical College and Research Institute, Chennai, TN, India
| | - Tessa Antony
- Microbiology, Sri Ramachandra Medical College and Research Institute, Chennai, TN, India
| | - Usha Rani Godla
- Obstetrics and Gynaecology, Sri Ramachandra Medical College and Research Institute, Chennai, TN, India
| | - Emmanuel Bhaskar
- General Medicine, Sri Ramachandra Medical College and Research Institute, Chennai, TN, India
| |
Collapse
|
24
|
Reese KL, Pantel K, Smit DJ. Multibiomarker panels in liquid biopsy for early detection of pancreatic cancer - a comprehensive review. J Exp Clin Cancer Res 2024; 43:250. [PMID: 39218911 PMCID: PMC11367781 DOI: 10.1186/s13046-024-03166-w] [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: 03/18/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is frequently detected in late stages, which leads to limited therapeutic options and a dismal overall survival rate. To date, no robust method for the detection of early-stage PDAC that can be used for targeted screening approaches is available. Liquid biopsy allows the minimally invasive collection of body fluids (typically peripheral blood) and the subsequent analysis of circulating tumor cells or tumor-associated molecules such as nucleic acids, proteins, or metabolites that may be useful for the early diagnosis of PDAC. Single biomarkers may lack sensitivity and/or specificity to reliably detect PDAC, while combinations of these circulating biomarkers in multimarker panels may improve the sensitivity and specificity of blood test-based diagnosis. In this narrative review, we present an overview of different liquid biopsy biomarkers for the early diagnosis of PDAC and discuss the validity of multimarker panels.
Collapse
Affiliation(s)
- Kim-Lea Reese
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany.
| | - Daniel J Smit
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany.
| |
Collapse
|
25
|
Löhr JM, Vujasinovic M, Kartalis N, Osten P. Pancreatic incidentaloma: incidental findings from history towards the era of liquid biopsy. EGASTROENTEROLOGY 2024; 2:e100082. [PMID: 39944362 PMCID: PMC11770461 DOI: 10.1136/egastro-2024-100082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/15/2024] [Indexed: 01/11/2025]
Abstract
This report provides an overview of the most common diagnostic methods that bring to light incidental findings of pancreatic cancer. It reviews the impact of medical imaging and genetic assessment on the definitions of incidental findings and incidentaloma of the pancreas. For different diagnostic approaches (eg, MRI and CT) and for different affections (cysts/intraductal papillary mucinous neoplasia, solid lesions), specific guidelines have been proposed and some are established. Based on this, we summarise the differences between the traditional methods with those applied in the PANCAID project. Biomarkers, genetic predispositions, mutations and circulating tumour cells give rise to different levels of concern. The final part of the report discusses the risks and the opportunities associated with further diagnostic procedures and surgical interventions. From the ethical perspective, the most urging question is, can a screening based on liquid biopsy and blood samples open a gateway for the prevention of pancreatic cancer-even if morbidity and lethality of today's surgical interventions is still very high?
Collapse
Affiliation(s)
| | | | | | - Philipp Osten
- Department of Medical History and Medical Ethics, University of Hamburg, Hamburg, Germany
| |
Collapse
|
26
|
Karamya ZA, Kovács A, Illés D, Czakó B, Fazekas A, Farkas N, Hegyi P, Czakó L. Prevalence of autoimmune pancreatitis in pancreatic resection for suspected malignancy: a systematic review and meta-analysis. BMC Gastroenterol 2024; 24:278. [PMID: 39169289 PMCID: PMC11337777 DOI: 10.1186/s12876-024-03367-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND/OBJECTIVES Autoimmune pancreatitis (AIP) is a diagnosis-challenging disease that often mimics pancreatic malignancy. Pancreatic resection is considered to be a curative treatment for pancreatic ductal adenocarcinoma (PDAC). This meta-analysis aims to study the incidence of AIP in patients who have undergone pancreatic resection for clinical manifestation of cancer. METHODS A comprehensive search was conducted in three databases, PubMed, Embase and the Cochrane Library, using the terms 'autoimmune pancreatitis' and 'pancreatic resection' and supplemented by manual checks of reference lists in all retrieved articles. RESULTS Ten articles were included in the final analysis. 8917 pancreatic resections were performed because of a clinical suspicion of pancreatic cancer. AIP accounted for 140 cases (1.6%). Type 1 AIP comprised the majority of cases, representing 94% (132 cases), while type 2 AIP made up the remaining 6% (eight cases) after further classification. AIP accounted for almost 26% of all cases of benign diseases involving unnecessary surgery and was overrepresented in males in 70% of cases compared to 30% in females. The mean age for AIP patients was 59 years. Serum CA 19 - 9 levels were elevated in 23 out of 47 (49%) AIP patients, where higher levels were detected more frequently in patients with type 1 AIP (51%, 22 out of 43) than in those with type 2 AIP (25%, 1 out of 4). The sensitivity of IgG4 levels in type 1 AIP was low (43%, 21/49 patients). CONCLUSION Even with modern diagnostic methods, distinguishing between AIP and PDAC can still be challenging, thus potentially resulting in unnecessary surgical procedures in some cases. Serum CA 19 - 9 levels are not useful in distinguishing between AIP and PDAC. Work must thus be done to improve diagnostic methods and avoid unnecessary complicated surgery.
Collapse
Affiliation(s)
- Zain A Karamya
- Division of Gastroenterology, Department of Medicine, University of Szeged, Pf.: 427, Szeged, H-6701, Hungary
| | - Attila Kovács
- Department of Gastroenterology and Internal Medicine, Markusovszky Teaching Hospital, Szombathely, Hungary
| | - Dóra Illés
- Division of Gastroenterology, Department of Medicine, University of Szeged, Pf.: 427, Szeged, H-6701, Hungary
| | - Bálint Czakó
- Division of Gastroenterology, Department of Medicine, University of Szeged, Pf.: 427, Szeged, H-6701, Hungary
| | - Alíz Fazekas
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Nelli Farkas
- Institute of Bioanalysis, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Hegyi
- Division of Pancreatic Diseases, Heart and Vascular Centre, Semmelweis University, Budapest, Hungary
| | - László Czakó
- Division of Gastroenterology, Department of Medicine, University of Szeged, Pf.: 427, Szeged, H-6701, Hungary.
| |
Collapse
|
27
|
Treekitkarnmongkol W, Dai J, Liu S, Sankaran D, Nguyen T, Balasenthil S, Hurd MW, Chen M, Katayama H, Roy-Chowdhuri S, Calin GA, Brand RE, Lampe PD, Hu TY, Maitra A, Koay EJ, Killary AM, Sen S. Blood-Based microRNA Biomarker Signature of Early-Stage Pancreatic Ductal Adenocarcinoma With Lead-Time Trajectory in Prediagnostic Samples. GASTRO HEP ADVANCES 2024; 3:1098-1115. [PMID: 39529638 PMCID: PMC11550741 DOI: 10.1016/j.gastha.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/02/2024] [Indexed: 11/16/2024]
Abstract
Background and Aims Clinically validated biomarker of pancreatic ductal adenocarcinoma (PDAC), carbohydrate antigen 19-9 (CA19-9), has limited sensitivity and specificity for early-stage disease. Circulating miRNAs in plasma associated with cancer relevant pathways were developed as early detection biomarkers. Methods 2083 miRNAs in 15 μl of plasma from multicenter age-matched cohorts (N = 203: healthy controls, n = 46; pancreatitis controls, n = 36; diagnosed cases: n = 121) and a prediagnostic Prostate, Lung, Colorectal, and Ovarian age- and gender-matched cohort (N = 96; controls, n = 48; prediagnosed cases, n = 48) were interrogated. A three-miRNA biomarker signature was developed for early-stage PDAC. Results The three-miRNA signature (let-7i-5p, miR-130a-3p and miR-221-3p) detected PDAC from healthy controls independently (area under the curve [AUC] of stage I, II, I-IV = 0.970, 0.975, 0.974) and in combination with CA19-9 (AUC of stage I, II, I-IV = 1.000, 0.992, 0.995). It also discriminated chronic pancreatitis (AUC of stage I, II, I-IV = 0.932, 0.931, 0.929), improving performance of CA19-9 alone (AUC of stage I, II, I-IV = 0.763, 0.701, 0.735) in combination (AUC of stage I, II, I-IV = 0.971, 0.943, 0.951). Blinded validation in prediagnostic Prostate, Lung, Colorectal, and Ovarian cohort revealed lead-time trajectory increase in AUC from 0.702 to 0.729 to 0.757 at twelve-, six-, and three-months before PDAC diagnosis, respectively. The signature also helped stratification of patients with different circulating tumor DNA and imaging subtypes. Conclusion Plasma miRNAs associated with oncogenic pathways may serve as PDAC early detection biomarkers.
Collapse
Affiliation(s)
- Warapen Treekitkarnmongkol
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jianliang Dai
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Suyu Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Deivendran Sankaran
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tristian Nguyen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Seetharaman Balasenthil
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark W. Hurd
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Meng Chen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hiroshi Katayama
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sinchita Roy-Chowdhuri
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - George A. Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Randall E. Brand
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Paul D. Lampe
- Translation Research Program, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Tony Y. Hu
- Department of Molecular & Cellular Biology, Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana
| | - Anirban Maitra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eugene J. Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ann M. Killary
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Subrata Sen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
28
|
Robles-Medranda C, Del Valle R, Puga-Tejada M, Arevalo-Mora M, Cunto D, Egas-Izquierdo M, Estrada-Guevara L, Bunces-Orellana O, Moreno-Zambrano D, Alcivar-Vasquez J, Alvarado-Escobar H, Merfea RC, Barreto-Perez J, Rodriguez J, Calle-Loffredo D, Pitanga-Lukashok H, Baquerizo-Burgos J, Tabacelia D. Assessing EUS-guided radiofrequency ablation in unresectable pancreatic ductal adenocarcinoma: a single-center historic cohort study. Gastrointest Endosc 2024; 100:250-258. [PMID: 38518978 DOI: 10.1016/j.gie.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/16/2024] [Accepted: 03/17/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND AND AIMS EUS-guided radiofrequency ablation (EUS-RFA) has emerged as an alternative for the local treatment of unresectable pancreatic ductal adenocarcinoma (PDAC). We assessed the feasibility and safety of EUS-RFA in patients with unresectable PDAC. METHODS This study followed an historic cohort compounded by locally advanced (LA-) and metastatic (m)PDAC-naïve patients who underwent EUS-RFA between October 2019 and March 2022. EUS-RFA was performed with a 19-gauge needle electrode with a 10-mm active tip for energy delivery. Study primary endpoints were feasibility, safety, and clinical follow-up, whereas secondary endpoints were performance status (PS), local control, and overall survival (OS). RESULTS Twenty-six patients were selected: 15 with locally advanced pancreatic duct adenocarcinoma (LA-PDAC) and 11 with metastatic pancreatic duct adenocarcinoma (mPDAC). Technical success was achieved in all patients with no major adverse events. Six months after EUS-RFA, OS was seen in 11 of 26 patients (42.3%), with significant PS improvement (P = .03). Local control was achieved, with tumor reduction from 39.5 mm to 26 mm (P = .04). A post-treatment hypodense necrotic area was observed at the 6-month follow-up in 11 of 11 patients who were still alive. Metastatic disease was a significant factor for worsening OS (hazard ratio, 5.021; 95% confidence interval, 1.589-15.87; P = .004). CONCLUSIONS EUS-RFA for the treatment of pancreatic adenocarcinoma is a minimally invasive and safe technique that may have an important role as targeted therapy for local treatment of unresectable cases and as an alternative for poor surgical candidates. Also, RFA may play a role in downstaging cancer with a potential increase in OS in nonmetastatic cases. Large prospective cohorts are required to evaluate this technique in clinical practice.
Collapse
Affiliation(s)
| | - Raquel Del Valle
- Instituto Ecuatoriano de Enfermedades Digestivas (IECED), Guayaquil, Ecuador
| | - Miguel Puga-Tejada
- Instituto Ecuatoriano de Enfermedades Digestivas (IECED), Guayaquil, Ecuador
| | - Martha Arevalo-Mora
- Instituto Ecuatoriano de Enfermedades Digestivas (IECED), Guayaquil, Ecuador
| | - Domenica Cunto
- Instituto Ecuatoriano de Enfermedades Digestivas (IECED), Guayaquil, Ecuador
| | | | | | | | | | | | | | - Ruxandra C Merfea
- Instituto Ecuatoriano de Enfermedades Digestivas (IECED), Guayaquil, Ecuador
| | | | - Jorge Rodriguez
- Instituto Ecuatoriano de Enfermedades Digestivas (IECED), Guayaquil, Ecuador
| | | | | | | | - Daniela Tabacelia
- Elias Emergency University Hospital, Bucharest, Romania; Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.
| |
Collapse
|
29
|
Bogdanski AM, van Hooft JE, Boekestijn B, Bonsing BA, Wasser MNJM, Klatte DCF, van Leerdam ME. Aspects and outcomes of surveillance for individuals at high-risk of pancreatic cancer. Fam Cancer 2024; 23:323-339. [PMID: 38619782 PMCID: PMC11255004 DOI: 10.1007/s10689-024-00368-1] [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: 01/05/2024] [Accepted: 02/24/2024] [Indexed: 04/16/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related deaths and is associated with a poor prognosis. The majority of these cancers are detected at a late stage, contributing to the bad prognosis. This underscores the need for novel, enhanced early detection strategies to improve the outcomes. While population-based screening is not recommended due to the relatively low incidence of PDAC, surveillance is recommended for individuals at high risk for PDAC due to their increased incidence of the disease. However, the outcomes of pancreatic cancer surveillance in high-risk individuals are not sorted out yet. In this review, we will address the identification of individuals at high risk for PDAC, discuss the objectives and targets of surveillance, outline how surveillance programs are organized, summarize the outcomes of high-risk individuals undergoing pancreatic cancer surveillance, and conclude with a future perspective on pancreatic cancer surveillance and novel developments.
Collapse
Affiliation(s)
- Aleksander M Bogdanski
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Jeanin E van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Bas Boekestijn
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin N J M Wasser
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Derk C F Klatte
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Reddy KS, Pandiaraj IP, Gaur A, Varatharajan S. Serum tumor markers: Can they clinically implicate in type 2 diabetes mellitus? World J Diabetes 2024; 15:1648-1650. [PMID: 39099811 PMCID: PMC11292342 DOI: 10.4239/wjd.v15.i7.1648] [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] [Received: 03/18/2024] [Revised: 05/08/2024] [Accepted: 05/24/2024] [Indexed: 07/08/2024] Open
Abstract
"Serum tumor markers expression (CA19-9, CA242, and CEA) and its clinical implications in type 2 diabetes mellitus" authored by Meng and Shi presents an observational case-control study investigating the correlation between tumor markers and type 2 diabetes mellitus (T2DM). The study explores the diagnostic accuracy of tumor markers, particularly cancer antigen 19-9 (CA19-9), CA242, and carcinoembryonic antigen, in poorly controlled T2DM patients with hemoglobin A1c levels exceeding 9%, employing receiver operating characteristic curve analysis. Though study offers valuable insights into the potential utility of tumor markers in clinical practice, caution is advised regarding routine tumor marker testing due to challenges such as limited availability and cost. Additionally, the study overlooks potential confounding factors like smoking and alcohol consumption. Variations in CA19-9 and CA242 expression underscore the complex interplay between tumor markers and systemic diseases, warranting further investigation into their diagnostic and prognostic implications. While Meng and Shi represent a significant contribution to the field, more extensive research is needed to fully elucidate the role of tumor markers in diabetes management and beyond.
Collapse
Affiliation(s)
- Kotha Sugunakar Reddy
- Department of General Medicine, All India Institute of Medical Sciences, Hyderabad 508126, Telangana, India
| | - Ilakkiya Priya Pandiaraj
- Department of General Medicine, All India Institute of Medical Sciences, Hyderabad 508126, Telangana, India
| | - Archana Gaur
- Department of Physiology, All India Institute of Medical Sciences, Hyderabad 508126, Telangana, India
| | - Sakthivadivel Varatharajan
- Department of General Medicine, All India Institute of Medical Sciences, Hyderabad 508126, Telangana, India
| |
Collapse
|
31
|
Rodriguez E, Zwart ES, Affandi AA, Verhoeff J, de Kok M, Boyd LNC, Meijer LL, Le Large TYS, Olesek K, Giovannetti E, García-Vallejo JJ, Mebius RE, van Kooyk Y, Kazemier G. In-depth immune profiling of peripheral blood mononuclear cells in patients with pancreatic ductal adenocarcinoma reveals discriminative immune subpopulations. Cancer Sci 2024; 115:2170-2183. [PMID: 38686549 PMCID: PMC11247553 DOI: 10.1111/cas.16147] [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: 09/16/2023] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 05/02/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis with a 5-year survival of less than 10%. More knowledge of the immune response developed in patients with PDAC is pivotal to develop better combination immune therapies to improve clinical outcome. In this study, we used mass cytometry time-of-flight to undertake an in-depth characterization of PBMCs from patients with PDAC and examine the differences with healthy controls and patients with benign diseases of the biliary system or pancreas. Peripheral blood mononuclear cells from patients with PDAC or benign disease are characterized by the increase of pro-inflammatory cells, as CD86+ classical monocytes and memory T cells expressing CCR6+ and CXCR3+, associated with T helper 1 (Th1) and Th17 immune responses, respectively. However, PBMCs from patients with PDAC present also an increase of CD39+ regulatory T cells and CCR4+CCR6-CXCR3- memory T cells, suggesting Th2 and regulatory responses. Concluding, our results show PDAC develops a multifaceted immunity, where a proinflammatory component is accompanied by regulatory responses, which could inhibit potential antitumor mechanisms.
Collapse
Affiliation(s)
- Ernesto Rodriguez
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Eline S Zwart
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
| | - Alsya A Affandi
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Jan Verhoeff
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Mike de Kok
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
| | - Lenka N C Boyd
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
| | - Laura L Meijer
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
| | - Tessa Y S Le Large
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
| | - Katarzyna Olesek
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Juan J García-Vallejo
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Geert Kazemier
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Kremser M, Weiss N, Kaufmann-Stoeck A, Vierbaum L, Schmitz A, Schellenberg I, Holdenrieder S. Longitudinal evaluation of external quality assessment results for CA 15-3, CA 19-9, and CA 125. Front Mol Biosci 2024; 11:1401619. [PMID: 38966130 PMCID: PMC11222321 DOI: 10.3389/fmolb.2024.1401619] [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: 03/15/2024] [Accepted: 05/30/2024] [Indexed: 07/06/2024] Open
Abstract
Background Tumor markers are established laboratory tools that help to diagnose, estimate prognosis, and monitor the course of cancer. For meaningful decision-making in patient care, it is essential that methods and analytical platforms demonstrate high sensitivity, specificity, precision, and comparability. Regular participation at external quality assessment (EQA) schemes is mandatory for laboratories. Here, a longitudinal evaluation of EQA data was performed to assess the performance of tumor marker assays over time. Methods Longitudinal data of the cancer antigens (CA) 15-3 (n = 5,492), CA 19-9 (n = 6,802), and CA 125 (n = 5,362) from 14 INSTAND EQAs conducted between 2019 and 2023 were evaluated. A median of 197, 244 and 191 laboratories participated at the EQAs for CA 15-3, CA 19-9 and CA 125, respectively. Data evaluation encompasses intra- and inter-manufacturer specific variations over time, assay precision, and adherence to the EQA limits of ±24% for CA 15-3, ±27% for CA 19-9 and ±36% for CA 125. Results The study showed median manufacturer-dependent differences of up to 107% for CA 15-3, 99% for CA 125, and even 549% for CA 19-9 between the highest and the lowest methods over the studied period. Regarding the normalized median of all methods, the values of the most deviant methods were 0.42 for CA 15-3, 7.61 for CA 19-9, and 1.82 for CA 125. Intra-manufacturer variability was generally low, with median coefficients of variation (CV) below 10%. As the methods were evaluated according to method-specific consensus values, most participants passed the EQAs within the acceptance criteria. When the criteria were consistently set at 24%, the central 90% of participants passed the EQAs in 78.6%-100% for CA 15-3 (with exception of AX), 89.3%-100% for CA 125, and 64.3%-100% for CA 19-9. Conclusion While intra-method precision of most analytical platforms is acceptable for all three tumor markers, considerable inter-method variability was observed over the whole studied period demonstrating the necessity for better standardization and harmonization of the methods, development of international reference materials, and comprehensive commutability studies with patient samples.
Collapse
Affiliation(s)
- Marcel Kremser
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Duesseldorf, Germany
| | - Nathalie Weiss
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Duesseldorf, Germany
| | - Anne Kaufmann-Stoeck
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Duesseldorf, Germany
| | - Laura Vierbaum
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Duesseldorf, Germany
| | - Arthur Schmitz
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Duesseldorf, Germany
| | - Ingo Schellenberg
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Duesseldorf, Germany
- Center of Life Sciences, Institute of Bioanalytical Sciences (IBAS), Anhalt University of Applied Sciences, Bernburg, Germany
| | - Stefan Holdenrieder
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Duesseldorf, Germany
- Munich Biomarker Research Center, Institute of Laboratory Medicine, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| |
Collapse
|
33
|
Mihailović J, Roganović J, Starčević I, Nikolić I, Prvulović Bunović N, Nikin Z. Diagnostic Performance of F-18 FDG PET/CT in the Detection of Recurrent Colorectal Cancer: Correlation with Biochemical Markers and Conventional Imaging Modalities. J Clin Med 2024; 13:3602. [PMID: 38930131 PMCID: PMC11204678 DOI: 10.3390/jcm13123602] [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: 04/30/2024] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Background/Objectives: Although the role of PET/CT imaging is well established in oncology, its diagnostic value in routine monitoring for recurrent colorectal cancer (CRC) is still controversial. The aim was to evaluate the diagnostic value of F-18 FDG PET/CT in detecting recurrent CRC in correlation with CEA, CA 19-9 levels, and conventional imaging modalities (CIM). Methods: Between 2009 and 2023, a retrospective study was performed including 134 CRC patients referred for PET/CT imaging on the suspicion of recurrence, based on elevated CEA and/or CA 19-9 and/or equivocal CIM findings. According to our institution's Tumor Board CRC protocol, after the initial treatment, which was dependent on the TNM stage (neoadjuvant therapy, primary resection, or adjuvant treatment), patients underwent a standard 5-year surveillance including CEA and CA 19-9 measurements, CIM, and colonoscopy, every six months. The statistics, including univariate and multivariate analyses were conducted using the IBM SPSS 20.0 statistical software. p-values < 0.05 were considered statistically significant. Results: Recurrent CRC was confirmed in 54/134 (40.3%) patients with elevated tumor markers. PET/CT showed high diagnostic performance in detecting recurrent CRC with sensitivity, specificity, PPV, NPV, and accuracy of 94.4%, 82.5%, 78.5%, 95.7%, and 87.3%, respectively. The CEA showed a high sensitivity of 98.1% but both low specificity and accuracy of 15% and 48.5%, respectively. The sensitivity, specificity, and accuracy for CA 19-9 and CIM for diagnosis of CRC recurrence were 44.4%, 67.5%, 58.2%, and 51.9%, 98.8%, 79.9%, respectively. The AUC for PET/CT, elevated CEA levels, CIM, and elevated CA 19-9 levels was 0.885 (95% CI: 0.824-0.946; p < 0.001), 0.844 (95% CI: 0.772-0.916; p < 0.001), 0.753 (95% CI: 0.612-0.844; p < 0.001), and 0.547 (95% CI: 0.442-0.652; p = 0.358), respectively. Univariate analysis showed that both PET/CT and CIM positive results were highly associated with CRC recurrence (p < 0.001 and p < 0.001, respectively). At the same time, gender, mucinous tumor type, presence of initial lymph node metastasis (N+), and presence of initial distant metastasis (M+) had no significance (p = 0.211, p = 0.158, p = 0.583, and p = 0.201, respectively). Our multivariate analysis showed that independent predictors for CRC recurrence are positive PET/CT scans (p < 0.001), positive CIM results (p = 0.001), and elevated CA 19-9 levels (p = 0.023). Although CA 19-9 was not detected as a statistically significant predictor in the univariate analysis (p = 0.358), in a multivariate analysis it was recognized as a significant predicting factor in detecting the CRC recurrence (p = 0.023). Conclusions: F-18 FDG PET/CT showed high diagnostic efficacy in CRC recurrence detection, in correlation with CEA levels, CA 19-9 levels, and CIM. This imaging modality should be routinely integrated into the post-operative follow-op in patients with elevated tumor markers.
Collapse
Affiliation(s)
- Jasna Mihailović
- Department of Nuclear Medicine, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia;
- Division of Nuclear Medicine, Oncology Institute of Vojvodina, Put dr Goldmana 4, 21204 Sremska Kamenica, Serbia; (J.R.); (I.S.)
| | - Jelena Roganović
- Division of Nuclear Medicine, Oncology Institute of Vojvodina, Put dr Goldmana 4, 21204 Sremska Kamenica, Serbia; (J.R.); (I.S.)
| | - Ivana Starčević
- Division of Nuclear Medicine, Oncology Institute of Vojvodina, Put dr Goldmana 4, 21204 Sremska Kamenica, Serbia; (J.R.); (I.S.)
| | - Ivan Nikolić
- Department of Internal Medicine, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia;
- Clinic for Medical Oncology, Oncology Institute of Vojvodina, Put dr Goldmana 4, 21204 Sremska Kamenica, Serbia
| | - Nataša Prvulović Bunović
- Department of Nuclear Medicine, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia;
- Centre for Diagnostic Imaging, Oncology Institute of Vojvodina, Put dr Goldmana 4, 21204 Sremska Kamenica, Serbia
| | - Zoran Nikin
- Department for Pathoanatomical Diagnostics, Oncology Institute of Vojvodina, Put dr Goldmana 4, 21204 Sremska Kamenica, Serbia;
| |
Collapse
|
34
|
Dişçi E, Peksöz R, Laloğlu E, Yıldırgan Mİ, Albayrak Y, Şirin MA, Ağırman E, Atamanalp SS. The Role of Serum Dickkopf1 and CKAP4 Levels in Diagnosing Colorectal Cancer and Measuring the Disease Severity: A Prospective Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:933. [PMID: 38929550 PMCID: PMC11205388 DOI: 10.3390/medicina60060933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/11/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024]
Abstract
Background and Objective: Colorectal cancer (CRC) is among the most common types of cancer. Although the disease is treatable in its early stages, five-year survival falls below 20% in the later stages. CEA and CA19-9 are tumor markers used in the diagnosis and follow-up of the disease in clinical practice; however, their diagnostic effectiveness is insufficient. Therefore, the identification of biomarkers that can be easily studied from serum and can diagnose CRC and determine its severity is highly important. In this context, dickkopf1 (DKK1) and cytoskeleton-associated protein 4 (CKAP4) are both promising biomarkers. Materials and Methods: Serum DKK1 and CKAP4 levels were measured in 55 patients with CRC and 40 healthy controls. The patients with CRC were divided into groups based on pathological stages and histological differentiation. The serum levels of both proteins in patients with CRC were measured preoperatively and 10 and 30 days postoperatively. Results: Serum DKK1 and CKAP4 were significantly higher in the CRC group than in the healthy controls (p < 0.05). Serum levels of both proteins rose in line with the disease stage and grade but decreased following surgical resection. A positive correlation was observed between tumor diameter and protein blood levels. The diagnostic efficacy of DKK1 and CKAP4 in CRC (approximately 95%) was higher than that of markers such as CEA and CA19-9. Conclusions: The DKK1 and CKAP4 serum values of patients with CRC are promising biomarkers. They can potentially be used in CRC management, namely, in the diagnosis and treatment of tumor response access and in tumor aggressiveness prediction.
Collapse
Affiliation(s)
- Esra Dişçi
- Department of General Surgery, Faculty of Medicine, Atatürk University, Erzurum 25240, Turkey; (E.D.); (R.P.); (M.İ.Y.)
| | - Rıfat Peksöz
- Department of General Surgery, Faculty of Medicine, Atatürk University, Erzurum 25240, Turkey; (E.D.); (R.P.); (M.İ.Y.)
| | - Esra Laloğlu
- Department Medical Biochemistry, Faculty of Medicine, Atatürk University, Erzurum 25240, Turkey
| | - Mehmet İlhan Yıldırgan
- Department of General Surgery, Faculty of Medicine, Atatürk University, Erzurum 25240, Turkey; (E.D.); (R.P.); (M.İ.Y.)
| | - Yavuz Albayrak
- Department of General Surgery, Faculty of Medicine, Atatürk University, Erzurum 25240, Turkey; (E.D.); (R.P.); (M.İ.Y.)
| | - Mehmet Akif Şirin
- Department of General Surgery, Faculty of Medicine, Atatürk University, Erzurum 25240, Turkey; (E.D.); (R.P.); (M.İ.Y.)
| | - Enes Ağırman
- Department of General Surgery, Erzurum City Hospital, Erzurum 25240, Turkey
| | - Sabri Selçuk Atamanalp
- Department of General Surgery, Faculty of Medicine, Atatürk University, Erzurum 25240, Turkey; (E.D.); (R.P.); (M.İ.Y.)
| |
Collapse
|
35
|
Chen X, Hu X, Liu T. Development of liquid biopsy in detection and screening of pancreatic cancer. Front Oncol 2024; 14:1415260. [PMID: 38887233 PMCID: PMC11180763 DOI: 10.3389/fonc.2024.1415260] [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: 04/12/2024] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
Pancreatic cancer is a highly lethal malignant tumor, which has the characteristics of occult onset, low early diagnosis rate, rapid development and poor prognosis. The reason for the high mortality is partly that pancreatic cancer is usually found in the late stage and missed the best opportunity for surgical resection. As a promising detection technology, liquid biopsy has the advantages of non-invasive, real-time and repeatable. In recent years, the continuous development of liquid biopsy has provided a new way for the detection and screening of pancreatic cancer. The update of biomarkers and detection tools has promoted the development of liquid biopsy. Circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), circulating tumor RNA (ctRNA) and extracellular vesicles (EVs) provide many biomarkers for liquid biopsy of pancreatic cancer, and screening tools around them have also been developed. This review aims to report the application of liquid biopsy technology in the detection of pancreatic cancer patients, mainly introduces the biomarkers and some newly developed tools and platforms. We have also considered whether liquid biopsy technology can replace traditional tissue biopsy and the challenges it faces.
Collapse
Affiliation(s)
- Xiangcheng Chen
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinyi Hu
- School of The First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tiancai Liu
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
36
|
Kramer RJ, Shi C, Moris D, Allen PJ. Benign Etiology for High-Risk Intraductal Papillary Mucinous Neoplasm: A Case Report and Literature Review. Cureus 2024; 16:e62054. [PMID: 38989360 PMCID: PMC11234795 DOI: 10.7759/cureus.62054] [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] [Accepted: 06/05/2024] [Indexed: 07/12/2024] Open
Abstract
Intraductal papillary mucinous neoplasms are relatively common and entail a variable risk of malignant potential. The Fukuoka guidelines present criteria for the risk of malignant transformation and are used for risk stratification and treatment decision-making. However, these guidelines entail some fallibility with limited sensitivity and specificity. In this case, we present an individual who had many of the hallmarks of malignant transformation but was found to have no evidence of malignancy or high-grade dysplasia. We discuss the suspected etiology of this individual's condition and how it might arise in others, as well as a brief review of the literature on risk factors in intraductal papillary mucinous neoplasms.
Collapse
Affiliation(s)
- Ryan J Kramer
- School of Medicine, Duke University School of Medicine, Durham, USA
| | - Chanjuan Shi
- Department of Pathology, Duke University Medical Center, Durham, USA
| | - Dimitrios Moris
- Department of Surgery, Duke University Medical Center, Durham, USA
| | - Peter J Allen
- Department of Surgical Oncology, Duke University Medical Center, Durham, USA
| |
Collapse
|
37
|
Pendry SD, Singhal N, Neo E, Foreman D, Winter JM. Elevation of the tumor marker CA19-9 in a pancreatic cancer survivor with benign prostatic hyperplasia: A clinical case report. Clin Case Rep 2024; 12:e8929. [PMID: 38799519 PMCID: PMC11126644 DOI: 10.1002/ccr3.8929] [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: 03/12/2024] [Revised: 04/24/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024] Open
Abstract
Serum carbohydrate antigen 19-9 (CA19-9) is used for recurrence surveillance in patients with resected pancreatic ductal adenocarcinoma (PDAC). This report describes the association of increasing CA19-9 in a male PDAC survivor with presence of prostatic hyperplasia. Unexplained elevation of CA19-9 in male PDAC survivors might be attributable to benign prostatic conditions.
Collapse
Affiliation(s)
- Steve D. Pendry
- Flinders Health and Medical ResearchCollege of Medicine and Public Health Flinders UniversityBedford ParkSouth AustraliaAustralia
| | - Nimit Singhal
- Cancer Centre, Royal Adelaide Hospital and School of MedicineThe University of AdelaideAdelaideSouth AustraliaAustralia
| | - Eu‐Ling Neo
- Hepatopancreatobiliary Surgical UnitFlinders Medical CentreBedford ParkSouth AustraliaAustralia
- Hepatobiliary Surgical UnitRoyal Adelaide HospitalAdelaideSouth AustraliaAustralia
| | - Darren Foreman
- College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Jean M. Winter
- Flinders Health and Medical ResearchCollege of Medicine and Public Health Flinders UniversityBedford ParkSouth AustraliaAustralia
| |
Collapse
|
38
|
Cotchim S, Kongkaew S, Thavarungkul P, Kanatharana P, Limbut W. A dual-electrode label-free immunosensor based on in situ prepared Au-MoO 3-Chi/porous graphene nanoparticles for point-of-care detection of cholangiocarcinoma. Talanta 2024; 272:125755. [PMID: 38364561 DOI: 10.1016/j.talanta.2024.125755] [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: 10/28/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/18/2024]
Abstract
A novel label-free electrochemical immunosensor was prepared for the detection of carbohydrate antigen 19-9 (CA19-9) and carcinoembryonic antigen (CEA) as biomarkers of cholangiocarcinoma (CCA). A nanocomposite of gold nanoparticles, molybdenum trioxide, and chitosan (Au-MoO3-Chi) was layer-by-layer assembled on the porous graphene (PG) modified a dual screen-printed electrode using a self-assembling technique, which increased surface area and conductivity and enhanced the adsorption of immobilized antibodies. The stepwise self-assembling procedure of the modified electrode was further characterized morphologically and functionally. The electroanalytical detection of biomarkers was based on the interaction between the antibody and antigen of each marker via linear sweep voltammetry using ferrocyanide/ferricyanide as an electrochemical redox indicator. Under optimized conditions, the fabricated immunosensor showed linear relationships between current change (ΔI) and antigen concentrations in two ranges: 0.0025-0.1 U mL-1 and 0.1-1.0 U mL-1 for CA19-9, and 0.001-0.01 ng mL-1 and 0.01-1.0 ng mL-1 for CEA. The limits of detection (LOD) were 1.0 mU mL-1 for CA19-9 and 0.5 pg mL-1 for CEA. Limits of quantitation (LOQ) were 3.3 mU mL-1 for CA19-9 and 1.6 pg mL-1 for CEA. The selectivity of the developed immunosensor was tested on mixtures of antigens and was then successfully applied to determine CA19-9 and CEA in human serum samples, producing satisfactory results consistent with the clinical method.
Collapse
Affiliation(s)
- Suparat Cotchim
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Division of Physical Science, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Supatinee Kongkaew
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Division of Physical Science, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Panote Thavarungkul
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Division of Physical Science, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Proespichaya Kanatharana
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Division of Physical Science, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Warakorn Limbut
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Forensic Science Innovation and Service Center, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand.
| |
Collapse
|
39
|
Liu XY, Wang XH. Effect of glycotoxicity and lipotoxicity on carbohydrate antigen 19 - 9 in the patients with diabetes. BMC Endocr Disord 2024; 24:51. [PMID: 38654232 PMCID: PMC11040910 DOI: 10.1186/s12902-024-01578-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
OBJECTIVES In comparison to the subjects without diabetes, a greater concentration of serum carbohydrate antigen 19 - 9 (CA 19 - 9) was observed in the subjects with diabetes. Nevertheless, since the occurrence of abnormal CA 19 - 9 is not widespread among the whole diabetic population, this phenomenon has not attracted enough attention. The prevalence of abnormal CA 19 - 9 in hospitalized patients with diabetes was the focus of our research. METHOD A total of 385 subjects with diabetes and 200 controls were enrolled and all had been tested the CA19-9 levels. Cases of cancers were excluded through examination and followup for 1 year. RESULTS We found that the rate of patients with abnormal CA19-9 level was 8.3%. The rate of patients with abnormal CA19-9 level was 14.0% in the HbA1c ≥ 9% group, and 3.0% in the HbA1c < 9% group, 2.5% in the control group. There was no significant difference in the HbA1c < 9% group and the control group. A significant correlation between serum CA19-9 and both HbA1c and total cholesterol was observed, yet no difference in CRP level was observed between subjects with normal CA19-9 level and subjects with abnormal CA19-9 level. However, a significant difference in fasting C-peptide levels was observed between the two groups, p = 0.039. CONCLUSION The percentage of patients with diabetes exhibiting elevated CA19-9 level is 14% in the HbA1c ≥ 9% diabetic patients, much higher than expected. The underlying mechanism may be related to islet injury caused by glycotoxicity and lipotoxicity. STRENGTHS AND LIMITATIONS OF THE STUDY We studied the rate of hospitalized diabetic patients with elevated CA 19 - 9 which were characterized with poorly controlled blood glucose. We found that the elevation of CA 19 - 9 was unexpectedly high in diabetic inpatients without development to cancer. The limitation of this study is that the underlying mechanism is not sufficiently studied.
Collapse
Affiliation(s)
- Xi-Yu Liu
- The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China.
| | - Xiao-Hong Wang
- The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| |
Collapse
|
40
|
Ohno E, Balduzzi A, Hijioka S, De Pastena M, Marchegiani G, Kato H, Takenaka M, Haba S, Salvia R. Association of high-risk stigmata and worrisome features with advanced neoplasia in intraductal papillary mucinous neoplasms (IPMN): A systematic review. Pancreatology 2024; 24:48-61. [PMID: 38161091 DOI: 10.1016/j.pan.2023.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/24/2023] [Accepted: 12/03/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND This systematic review aimed to assess the diagnostic accuracy of the International Consensus Fukuoka Guidelines (ICG2017) in identifying high-risk lesions of Intraductal Papillary Mucinous Neoplasms (IPMNs). METHODS The ICG2017 revision committee conducted a comprehensive literature review to establish evidence-based statements on IPMNs. The review focused on articles examining the diagnostic value of imaging features (e.g., cyst or main pancreatic duct diameter), clinical symptoms associated with IPMN, and serum biomarkers. Five clinical questions regarding high-risk stigmata (HRS) and worrisome features (WF) in the ICG2017 guidelines were addressed. RESULTS A total of 210 articles were reviewed. The findings revealed a significant association between the presence of mural nodules ≥5 mm in diameter or solid components with contrast enhancement and the diagnosis of high-grade dysplasia or invasive carcinoma. Contrast-enhanced diagnostic tools, such as CT, MRI, or EUS, demonstrated the highest prediction rate and were recommended. Positive cytology was identified as an HRS, while symptoms like acute pancreatitis and cyst diameter growth ≥2.5 mm per year were considered WFs. The use of nomograms and multiple diagnostic factors was recommended for optimal IPMN management. CONCLUSIONS This systematic review provides evidence supporting the improved diagnostic accuracy of ICG2017 in identifying high-risk lesions of IPMN. The multidisciplinary incorporation of HRS and WF based on imaging findings and clinical symptoms is crucial. These findings should inform the revision of ICG2017, enhancing the evaluation and management of IPMN patients. By implementing these recommendations, clinicians can make more informed decisions, leading to better diagnosis and treatment outcomes for high-risk IPMN cases.
Collapse
Affiliation(s)
- Eizaburo Ohno
- Fujita Health University, Department of Gastroenterology and Hepatology, Japan; Nagoya University Hospital, Department of Gastroenterology and Hepatology, Japan
| | - Alberto Balduzzi
- The Pancreas Institute Verona, Unit of General and Pancreatic Surgery Department of Surgery, Dentistry, Paediatrics and Gynaecology University of Verona, Italy
| | - Susumu Hijioka
- National Cancer Center Hospital, Department of Hepatobiliary and Pancreatic Oncology, Japan
| | - Matteo De Pastena
- The Pancreas Institute Verona, Unit of General and Pancreatic Surgery Department of Surgery, Dentistry, Paediatrics and Gynaecology University of Verona, Italy
| | - Giovanni Marchegiani
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Unit, Department of Surgical, Oncological and Gastroenterological Sciences (DISCOG), University of Padua, Padua, Italy
| | - Hironari Kato
- Okayama University Hospital, Department of Gastroenterology and Hepatology, Japan
| | - Mamoru Takenaka
- Kindai University, Department of Gastroenterology and Hepatology, Japan
| | - Shin Haba
- Aichi Cancer Center Hospital, Department of Gastroenterology, Japan
| | - Roberto Salvia
- The Pancreas Institute Verona, Unit of General and Pancreatic Surgery Department of Surgery, Dentistry, Paediatrics and Gynaecology University of Verona, Italy.
| |
Collapse
|
41
|
Nooijen LE, van der Snee L, Ten Haaft B, Kazemier G, Klümpen HJ, Bridgewater J, Primrose J, Erdmann J. A critical appraisal of the potential benefit of post-operative structured follow-up after resection for biliary tract cancer. HPB (Oxford) 2024; 26:179-187. [PMID: 37891150 DOI: 10.1016/j.hpb.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/19/2023] [Accepted: 10/01/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND There is currently no evidence to support structured use of imaging or biomarkers during follow-up of patients after curative resection of biliary tract cancer (BTC). Besides, the influence of early detection of recurrence and subsequent start of palliative chemotherapy on overall survival remains unknown. The aim of this study is to describe and compare the results of two follow-up strategies. METHODS This retrospective multicenter cohort study compared patients from the Amsterdam UMC undergoing pragmatic clinical follow-up, to patients from the observational cohort of the BILCAP study undergoing structured follow-up. Primary outcome was overall survival. RESULTS A total of 315 patients were included n=91 pragmatic, n=224 structured follow-up). At median follow-up of 56.9 months, 189 (60%) patients were diagnosed with recurrence. After recurrence, more patients received palliative (chemo) therapy in the structured group (43% vs 75%, P<0.001). Median overall survival was lower in the pragmatic group (27.7 vs 39.1 months, P=0.003). Median overall survival of patients who actually received chemotherapy was comparable (27.2 vs 27.7 months, P=0.574). CONCLUSION This study describes the results of two follow-up strategies. Although these groups are biased, it is noted that after pragmatic follow-up fewer patients received palliative chemotherapy but that those who actually received chemotherapy had similar overall survival compared to patients undergoing structured follow-up.
Collapse
Affiliation(s)
- Lynn E Nooijen
- Amsterdam UMC, Location Vrije Universiteit Amsterdam, Department of Surgery, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| | - Lizzel van der Snee
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands; Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands
| | - Britte Ten Haaft
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands; Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands
| | - Geert Kazemier
- Amsterdam UMC, Location Vrije Universiteit Amsterdam, Department of Surgery, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| | - Heinz-Josef Klümpen
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands; Amsterdam UMC, Location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands
| | - John Bridgewater
- UCL Cancer Institute, Department of Medical Oncology, London, United Kingdom
| | - John Primrose
- University of Southampton, Department of Surgery, Southampton, United Kingdom
| | - Joris Erdmann
- Amsterdam UMC, Location Vrije Universiteit Amsterdam, Department of Surgery, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands.
| |
Collapse
|
42
|
Kafle A, Suttiprapa S. Current State of Knowledge on Blood and Tissue-Based Biomarkers for Opisthorchis viverrini-induced Cholangiocarcinoma: A Review of Prognostic, Predictive, and Diagnostic Markers. Asian Pac J Cancer Prev 2024; 25:25-41. [PMID: 38285765 PMCID: PMC10911713 DOI: 10.31557/apjcp.2024.25.1.25] [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] [Accepted: 01/19/2024] [Indexed: 01/31/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a prevalent cancer in Southeast Asia, with Opisthorchis viverrini (O.viverrini) infection being the primary risk factor. Most CCA cases in this region are diagnosed at advanced stages, leading to unfavorable prognoses. The development of stage-specific biomarkers for Opisthorchis viverrini-induced cholangiocarcinoma (Ov-CCA) holds crucial significance, as it facilitates early detection and timely administration of curative interventions, effectively mitigating the high morbidity and mortality rates associated with this disease in the Great Mekong region. Biomarkers are a promising approach for early detection, prognosis, and targeted treatment of CCA. Disease-specific biomarkers facilitate early detection and enable monitoring of therapy effectiveness, allowing for any necessary corrections. This review provides an overview of the potential O. viverrini-specific molecular biomarkers and important markers for diagnosing and monitoring Ov-CCA, discussing their prognostic, predictive, and diagnostic value. Despite the limited research in this domain, several potential biomarkers have been identified, encompassing both worm-induced and host-induced factors. This review offers a thorough examination of historical and contemporary progress in identifying biomarkers through multiomics techniques, along with their potential implications for early detection and treatment.
Collapse
Affiliation(s)
- Alok Kafle
- Tropical Medicine Graduate Program, Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Sutas Suttiprapa
- Tropical Medicine Graduate Program, Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.
- Tropical Disease Research Center, WHO Collaborating Centre for Research and Control of Opisthorchiasis, Khon Kaen University, Khon Kaen 40002, Thailand.
| |
Collapse
|
43
|
Trapé J, Fernández-Galán E, Auge JM, Carbonell-Prat M, Filella X, Miró-Cañís S, González-Fernández C. Factors influencing blood tumor marker concentrations in the absence of neoplasia. Tumour Biol 2024; 46:S35-S63. [PMID: 38517826 DOI: 10.3233/tub-220023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024] Open
Abstract
BACKGROUND Tumor markers (TMs) are a heterogeneous group of molecules used in the diagnosis, prognosis and follow-up of cancer patients. During neoplastic differentiation, cells can either directly synthesize or induce the synthesis of TMs, and the release of these molecules into the bloodstream allows their quantification in biological fluids. Although very small concentrations of TMs are usually present in the serum or plasma of healthy subjects, increased concentrations may also be found in the presence of benign diseases or due to technical interference, producing false positive results. MATERIAL AND METHODS AND RESULTS Our review analyses the causes of false positives described between January 1970 to February 2023 for the TMs most frequently used in clinical practice: α-fetoprotein (AFP), β2-microglobulin (β2-M), cancer antigen 15-3 (CA 15-3), cancer antigen CA 19-9 (CA 19-9), cancer antigen CA 72-4 (CA 72-4), cancer antigen 125 (CA 125), carcinoembryonic antigen (CEA), chromogranin A (CgA), choriogonadotropin (hCG), cytokeratin 19 fragment (CYFRA 21-1), neuron-specific enolase (NSE), human epididymis protein 4 (HE4), serum HER2 (sHER2), squamous cell carcinoma antigen (SCCA), protein induced by vitamin K absence-II (PIVKA-II), Pro-gastrin-releasing peptide (Pro-GRP), prostate-specific antigen (PSA), Protein S-100 (S-100) and thyroglobulin (Tg). A total of 247 references were included. CONCLUSIONS A better understanding of pathophysiological processes and other conditions that affect the concentration of TMs might improve the interpretation of results and their clinical application.
Collapse
Affiliation(s)
- Jaume Trapé
- Department of Laboratory Medicine, Althaia Xarxa Assistencial Universitària de Manresa, Manresa, Catalonia, Spain
- Tissue Repair and Regeneration Laboratory, Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central, Barcelona, Spain
- Faculty of Medicine, University of Vic - Central University of Catalonia, Vic, Spain
| | - Esther Fernández-Galán
- Department of Biochemistry and Molecular Genetics - Hospital Clinic de Barcelona, Barcelona, Spain
| | - Josep Maria Auge
- Department of Biochemistry and Molecular Genetics - Hospital Clinic de Barcelona, Barcelona, Spain
| | | | - Xavier Filella
- Department of Biochemistry and Molecular Genetics - Hospital Clinic de Barcelona, Barcelona, Spain
| | - Sílvia Miró-Cañís
- Laboratori d'Anàlisis Clíniques, CLILAB Diagnòstics, Vilafranca del Penedès, Spain
| | - Carolina González-Fernández
- Department of Laboratory Medicine, Althaia Xarxa Assistencial Universitària de Manresa, Manresa, Catalonia, Spain
- Gastrointestinal Oncology, Endoscopy and Surgery Research Group, Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central, Barcelona, Spain
| |
Collapse
|
44
|
Hung TH, Yeh CN, Hung JT, Wu CE, Lee CW, Yu J, Yu AL, Huang Y. Globo H ceramide is an independent prognostic marker for gallbladder cancer. Am J Cancer Res 2023; 13:4811-4821. [PMID: 37970342 PMCID: PMC10636676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/23/2023] [Indexed: 11/17/2023] Open
Abstract
In recent studies, there has been growing interest in developing cancer therapeutics targeting Globo H ceramide, which is considered as the most prevalent tumor-associated carbohydrate antigen in epithelial cancers. In this study, we aimed to evaluate the expression of Globo H and investigate its prognostic significance in gallbladder cancer (GBC). The tumor specimens and clinical characteristics of GBC patients were collected from the tumor bank and database of Chang Gung Memorial Hospital. Globo H in tumor specimens was detected by immunohistochemistry (IHC) and mass spectrometry analysis. Through data mining, it was discovered that FUT1 and FUT2, which are key enzymes involved in the biosynthesis of Globo H, were significantly up-regulated in human gallbladder cancer (GBC). Consistent with this finding, Globo H expression was detected in 86% (128 out of 149) of GBC specimens using immunohistochemical (IHC) staining. This was the highest frequency among Globo H expressing cancers. Patients with tumors exhibiting higher Globo H expression (H-score ≥ 80) demonstrated significantly shorter disease-free survival (DFS) and overall survival (OS) (P = 0.0001 and P = 0.0004, respectively). In a multivariable Cox regression analysis, elevated Globo H expression was identified as an independent unfavorable predictor for DFS and OS (hazard ratio: 2.29 and 2.32, respectively, P = 0.008 and 0.001) in primary GBC. Globo H is an independent prognostic marker for GBC.
Collapse
Affiliation(s)
- Tsai-Hsien Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at LinkouTaoyuan 333, Taiwan
| | - Chun-Nan Yeh
- Department of Surgery and Liver Research Center, Chang Gung Memorial Hospital, Chang Gung UniversityTaoyuan 333, Taiwan
| | - Jung-Tung Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at LinkouTaoyuan 333, Taiwan
| | - Chiao-En Wu
- Department of Hematology-Oncology, Chang Gung Memorial Hospital, Chang Gung UniversityTaoyuan 333, Taiwan
| | - Chien-Wei Lee
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at LinkouTaoyuan 333, Taiwan
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at LinkouTaoyuan 333, Taiwan
- Chang Gung UniversityTaoyuan 333, Taiwan
| | - Alice L. Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at LinkouTaoyuan 333, Taiwan
- Chang Gung UniversityTaoyuan 333, Taiwan
- Department of Pediatrics, University of California in San DiegoSan Diego, CA 92103, USA
| | - Yenlin Huang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at LinkouTaoyuan 333, Taiwan
- School of Medicine, National Tsing Hua UniversityHsinchu 300, Taiwan
- Department of Anatomic Pathology, Chang Gung Memorial Hospital at LinkouTaoyuan 333, Taiwan
| |
Collapse
|
45
|
Dbouk M, Abe T, Koi C, Ando Y, Saba H, Abou Diwan E, MacGregor-Das A, Blackford AL, Mocci E, Beierl K, Dbouk A, He J, Burkhart R, Lennon AM, Sokoll L, Canto MI, Eshleman JR, Goggins M. Diagnostic Performance of a Tumor Marker Gene Test to Personalize Serum CA19-9 Reference Ranges. Clin Cancer Res 2023; 29:4178-4185. [PMID: 37566230 PMCID: PMC10570677 DOI: 10.1158/1078-0432.ccr-23-0655] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/25/2023] [Accepted: 08/08/2023] [Indexed: 08/12/2023]
Abstract
PURPOSE CA19-9 synthesis is influenced by common variants in the fucosyltransferase (FUT) enzymes FUT3 and FUT2. We developed a clinical test to detect FUT variants, and evaluated its diagnostic performance for pancreatic ductal adenocarcinoma (PDAC). EXPERIMENTAL DESIGN A representative set of controls from the Cancer of the Pancreas Screening study was identified for each FUT functional group. Diagnostic sensitivity was determined first in a testing set of 234 PDAC cases, followed by a 134-case validation set, all of whom had undergone resection with curative intent without neoadjuvant therapy. Tumor marker gene testing was performed in the Johns Hopkins Molecular Diagnostics Laboratory. CA19-9 levels were measured in the Hopkins Clinical Chemistry lab. Receiver operating characteristic (ROC) curve analysis was used to evaluate the discriminative ability of CA19-9 alone versus with the gene test. RESULTS Applying the CA19-9 standard cutoff (<36 U/mL) to all 716 subjects yielded a 68.8% sensitivity in the test set of cases, 67.2% in the validation set, at 91.4% specificity. Applying 99th percentile cutoffs according to each individual's FUT group (3, 34.9, 41.8, and 89.2, for the FUT3-null, FUT-low, FUT-intermediate, and FUT-high groups, respectively) yielded a diagnostic sensitivity for CA19-9 in the first set of cases of 66.7%, 65.7% in the validation set, at 98.9% specificity. ROC analysis for CA19-9 alone yielded an AUC of 0.84; with the tumor marker gene test, AUC improved to 0.92 (P < 0.001). CONCLUSIONS Using a tumor marker gene test to personalize an individual's CA19-9 reference range significantly improves diagnostic accuracy.
Collapse
Affiliation(s)
- Mohamad Dbouk
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Toshiya Abe
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Chiho Koi
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Yohei Ando
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Helena Saba
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Elizabeth Abou Diwan
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Anne MacGregor-Das
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Amanda L. Blackford
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Evelina Mocci
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Katie Beierl
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Ali Dbouk
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Jin He
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Richard Burkhart
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Anne Marie Lennon
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Lori Sokoll
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Marcia Irene Canto
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - James R. Eshleman
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Michael Goggins
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| |
Collapse
|
46
|
McConnell A, Stoneman T, Hewlett S. Extraordinarily high serum CA 19-9 in setting of pancreatic necrosis and underlying pancreatic adenocarcinoma: a case report. J Surg Case Rep 2023; 2023:rjad550. [PMID: 37846412 PMCID: PMC10576995 DOI: 10.1093/jscr/rjad550] [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: 08/03/2023] [Accepted: 09/21/2023] [Indexed: 10/18/2023] Open
Abstract
Carbohydrate antigen (CA 19-9) is the most validated marker for both sensitivity and specificity of pancreatic adenocarcinoma used to aid diagnosis of symptomatic patients as well as to evaluate the progression or treatment of disease. Though higher levels of CA 19-9 tend to correlate with neoplastic disease, elevated levels are also often seen in patients with benign gastrointestinal diseases, such as obstructive jaundice and pancreatitis. We present a case of a 74-year-old male who was admitted for abdominal pain and worsening jaundice who was diagnosed with extensive pancreatic necrosis and an underlying invasive pancreatic adenocarcinoma whose serum level of CA 19-9 was found to be extraordinarily high.
Collapse
Affiliation(s)
- Ashlyn McConnell
- Department of Surgery, Princeton Baptist Medical Center, Birmingham, AL 35211, United States
| | - Tyler Stoneman
- Edward Via College of Osteopathic Medicine, Spartanburg, SC 29303, United States
| | - Stanley Hewlett
- Department of Surgery, Princeton Baptist Medical Center, Birmingham, AL 35211, United States
| |
Collapse
|
47
|
Freed IM, Kasi A, Fateru O, Hu M, Gonzalez P, Weatherington N, Pathak H, Hyter S, Sun W, Al-Rajabi R, Baranda J, Hupert ML, Chalise P, Godwin AK, A. Witek M, Soper SA. Circulating Tumor Cell Subpopulations Predict Treatment Outcome in Pancreatic Ductal Adenocarcinoma (PDAC) Patients. Cells 2023; 12:2266. [PMID: 37759489 PMCID: PMC10526802 DOI: 10.3390/cells12182266] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/06/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
There is a high clinical unmet need to improve outcomes for pancreatic ductal adenocarcinoma (PDAC) patients, either with the discovery of new therapies or biomarkers that can track response to treatment more efficiently than imaging. We report an innovative approach that will generate renewed interest in using circulating tumor cells (CTCs) to monitor treatment efficacy, which, in this case, used PDAC patients receiving an exploratory new therapy, poly ADP-ribose polymerase inhibitor (PARPi)-niraparib-as a case study. CTCs were enumerated from whole blood using a microfluidic approach that affinity captures epithelial and mesenchymal CTCs using anti-EpCAM and anti-FAPα monoclonal antibodies, respectively. These antibodies were poised on the surface of two separate microfluidic devices to discretely capture each subpopulation for interrogation. The isolated CTCs were enumerated using immunophenotyping to produce a numerical ratio consisting of the number of mesenchymal to epithelial CTCs (denoted "Φ"), which was used as an indicator of response to therapy, as determined using computed tomography (CT). A decreasing value of Φ during treatment was indicative of tumor response to the PARPi and was observed in 88% of the enrolled patients (n = 31). Changes in Φ during longitudinal testing were a better predictor of treatment response than the current standard CA19-9. We were able to differentiate between responders and non-responders using ΔΦ (p = 0.0093) with higher confidence than CA19-9 (p = 0.033). For CA19-9 non-producers, ΔΦ correctly predicted the outcome in 72% of the PDAC patients. Sequencing of the gDNA extracted from affinity-selected CTC subpopulations provided information that could be used for patient enrollment into the clinical trial based on their tumor mutational status in DNA repair genes.
Collapse
Affiliation(s)
- Ian M. Freed
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (I.M.F.); (O.F.); (M.H.); (P.G.); (N.W.); (M.A.W.)
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
| | - Anup Kasi
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (W.S.); (R.A.-R.); (J.B.); (P.C.)
| | - Oluwadamilola Fateru
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (I.M.F.); (O.F.); (M.H.); (P.G.); (N.W.); (M.A.W.)
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
| | - Mengjia Hu
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (I.M.F.); (O.F.); (M.H.); (P.G.); (N.W.); (M.A.W.)
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.P.); (S.H.)
- Department of Cancer Biology, The University of Kansas Medical Center, Cancer Center, Kansas City, KS 66160, USA
| | - Phasin Gonzalez
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (I.M.F.); (O.F.); (M.H.); (P.G.); (N.W.); (M.A.W.)
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
| | - Nyla Weatherington
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (I.M.F.); (O.F.); (M.H.); (P.G.); (N.W.); (M.A.W.)
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
| | - Harsh Pathak
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.P.); (S.H.)
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Stephen Hyter
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.P.); (S.H.)
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Weijing Sun
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (W.S.); (R.A.-R.); (J.B.); (P.C.)
| | - Raed Al-Rajabi
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (W.S.); (R.A.-R.); (J.B.); (P.C.)
| | - Joaquina Baranda
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (W.S.); (R.A.-R.); (J.B.); (P.C.)
| | | | - Prabhakar Chalise
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (W.S.); (R.A.-R.); (J.B.); (P.C.)
| | - Andrew K. Godwin
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.P.); (S.H.)
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Malgorzata A. Witek
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (I.M.F.); (O.F.); (M.H.); (P.G.); (N.W.); (M.A.W.)
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.P.); (S.H.)
| | - Steven A. Soper
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (I.M.F.); (O.F.); (M.H.); (P.G.); (N.W.); (M.A.W.)
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.P.); (S.H.)
- Department of Cancer Biology, The University of Kansas Medical Center, Cancer Center, Kansas City, KS 66160, USA
- BioFluidica, Inc., San Diego, CA 92121, USA;
- Bioengineering Program, The University of Kansas, Lawrence, KS 66045, USA
- Department of Mechanical Engineering, The University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
48
|
Levink IJM, Jaarsma SC, Koopmann BDM, van Riet PA, Overbeek KA, Meziani J, Sprij MLJA, Casadei R, Ingaldi C, Polkowski M, Engels MML, van der Waaij LA, Carrara S, Pando E, Vornhülz M, Honkoop P, Schoon EJ, Laukkarinen J, Bergmann JF, Rossi G, van Vilsteren FGI, van Berkel A, Tabone T, Schwartz MP, Tan ACITL, van Hooft JE, Quispel R, van Soest E, Czacko L, Bruno MJ, Cahen DL. The additive value of CA19.9 monitoring in a pancreatic cyst surveillance program. United European Gastroenterol J 2023; 11:601-611. [PMID: 37435855 PMCID: PMC10493362 DOI: 10.1002/ueg2.12422] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 05/05/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND Surveillance of pancreatic cysts focuses on the detection of (mostly morphologic) features warranting surgery. European guidelines consider elevated CA19.9 as a relative indication for surgery. We aimed to evaluate the role of CA19.9 monitoring for early detection and management in a cyst surveillance population. METHODS The PACYFIC-registry is a prospective collaboration that investigates the yield of pancreatic cyst surveillance performed at the discretion of the treating physician. We included participants for whom at least one serum CA19.9 value was determined by a minimum follow-up of 12 months. RESULTS Of 1865 PACYFIC participants, 685 met the inclusion criteria for this study (mean age 67 years, SD 10; 61% female). During a median follow-up of 25 months (IQR 24, 1966 visits), 29 participants developed high-grade dysplasia (HGD) or pancreatic cancer. At baseline, CA19.9 ranged from 1 to 591 kU/L (median 10 kU/L [IQR 14]), and was elevated (≥37 kU/L) in 64 participants (9%). During 191 of 1966 visits (10%), an elevated CA19.9 was detected, and these visits more often led to an intensified follow-up (42%) than those without an elevated CA19.9 (27%; p < 0.001). An elevated CA19.9 was the sole reason for surgery in five participants with benign disease (10%). The baseline CA19.9 value was (as continuous or dichotomous variable at the 37 kU/L threshold) not independently associated with HGD or pancreatic cancer development, whilst a CA19.9 of ≥ 133 kU/L was (HR 3.8, 95% CI 1.1-13, p = 0.03). CONCLUSIONS In this pancreatic cyst surveillance cohort, CA19.9 monitoring caused substantial harm by shortening surveillance intervals (and performance of unnecessary surgery). The current CA19.9 cutoff was not predictive of HGD and pancreatic cancer, whereas a higher cutoff may decrease false-positive values. The role of CA19.9 monitoring should be critically appraised prior to implementation in surveillance programs and guidelines.
Collapse
Affiliation(s)
- Iris J. M. Levink
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
| | - Sanne C. Jaarsma
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
| | - Brechtje D. M. Koopmann
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
- Department of Public HealthErasmus University Medical CenterRotterdamThe Netherlands
| | - Priscilla A. van Riet
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
| | - Kasper A. Overbeek
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
| | - Jihane Meziani
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
| | - Marloes L. J. A. Sprij
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
| | | | | | - Marcin Polkowski
- Department of Gastroenterology, Hepatology, and Clinical OncologyCenter of Postgraduate Medical EducationWarsawPoland
- Department of Oncological GastroenterologyThe Maria Sklodowska‐Curie National Research Institute of OncologyWarsawPoland
| | - Megan M. L. Engels
- Department of Gastroenterology & HepatologyMayo ClinicJacksonvilleFloridaUSA
- Department of Gastroenterology & HepatologyLeiden University Medical CenterLeidenThe Netherlands
| | | | - Silvia Carrara
- Department of GastroenterologyIRCCS Humanitas Research HospitalMilanItaly
| | - Elizabeth Pando
- Department of SurgeryVall d’Hebron Institute of ResearchBarcelonaSpain
| | - Marlies Vornhülz
- Department of Gastroenterology & HepatologyLudwig‐Maximilians‐University of MunichMunichGermany
| | - Pieter Honkoop
- Department of Gastroenterology & HepatologyAlbert Schweitzer HospitalDordrechtThe Netherlands
| | - Erik J. Schoon
- Department of Gastroenterology & HepatologyCatharina HospitalEindhovenThe Netherlands
| | | | - Jilling F. Bergmann
- Department of Gastroenterology & HepatologyHaga ZiekenhuisThe HagueThe Netherlands
| | - Gemma Rossi
- Pancreato‐Biliary Endoscopy and Endosonography DivisionPancreas Translational and Clinical Research CenterSan Raffaele Scientific Institute IRCCSVita Salute San Raffaele UniversityMilanItaly
| | | | | | - Trevor Tabone
- Department of Gastroenterology & HepatologyMater dei HospitalMsidaMalta
| | - Matthijs P. Schwartz
- Department of Gastroenterology & HepatologyMeander Medical CenterAmersfoortThe Netherlands
| | - Adriaan C. I. T. L. Tan
- Department of Gastroenterology & HepatologyCanisius Wilhelmina HospitalNijmegenThe Netherlands
| | - Jeanin E. van Hooft
- Department of Gastroenterology & HepatologyLeiden University Medical CenterLeidenThe Netherlands
- Department of Gastroenterology & HepatologyAmsterdam UMCAmsterdamThe Netherlands
| | - Rutger Quispel
- Department of Gastroenterology & HepatologyReinier de GraafDelftThe Netherlands
| | - Ellert van Soest
- Department of Gastroenterology & HepatologySpaarne GasthuisHaarlemThe Netherlands
| | - Laszlo Czacko
- Department of Gastroenterology & HepatologyUniversity of SzegedSzegedHungary
| | - Marco J. Bruno
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
| | - Djuna L. Cahen
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
- Department of Gastroenterology & HepatologyAmstellandAmstelveenThe Netherlands
| | | |
Collapse
|
49
|
Chiu YF, Liu TW, Shan YS, Chen JS, Li CP, Ho CL, Hsieh RK, Hwang TL, Chen LT, Ch'ang HJ. Carbohydrate Antigen 19-9 Response to Initial Adjuvant Chemotherapy Predicts Survival and Failure Pattern of Resected Pancreatic Adenocarcinoma but Not Which Patients Are Suited for Additional Adjuvant Chemoradiation Therapy: From a Prospective Randomized Study. Int J Radiat Oncol Biol Phys 2023; 117:74-86. [PMID: 37055279 DOI: 10.1016/j.ijrobp.2023.02.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 02/19/2023] [Accepted: 02/25/2023] [Indexed: 04/15/2023]
Abstract
PURPOSE The predictive value of carbohydrate antigen 19-9 (CA19-9) for adjuvant chemo(radiation) therapy of resected pancreatic adenocarcinoma (PDAC) is undefined. METHODS AND MATERIALS We analyzed CA19-9 levels in patients with resected PDAC in a prospective randomized trial of adjuvant chemotherapy with or without additional chemoradiation therapy (CRT). Patients with postoperative CA19-9 ≤92.5 U/mL and serum bilirubin ≤2 mg/dL were randomized to 2 arms: patients in 1 arm received 6 cycles of gemcitabine, whereas those in the other received 3 cycles of gemcitabine followed by CRT and another 3 cycles of gemcitabine. Serum CA19-9 was measured every 12 weeks. Those who had CA19-9 levels always <3 U/mL were excluded from the exploratory analysis. RESULTS One hundred forty-seven patients were enrolled in this randomized trial. Twenty-two patients with CA19-9 levels always ≤3 U/mL were excluded from the analysis. For the 125 participants, median overall survival (OS) and recurrence-free survival were 23.1 and 12.1 months, respectively, with no significant differences between the study arms. Postresection CA19-9 levels and, to a lesser extent, CA19-9 change predicted OS (P = .040 and .077, respectively). For the 89 patients who completed the initial 3 cycles of adjuvant gemcitabine, the CA19-9 response was significantly correlated with initial failure over the distant site (P = .023) and OS (P = .0022). Despite a trend of less initial failure over the locoregional area (P = .031), neither postoperative CA19-9 level nor CA19-9 response helped to select patients who might have a survival benefit from additional adjuvant CRT. CONCLUSIONS CA19-9 response to initial adjuvant gemcitabine predicts survival and distant failure of PDAC after resection; however, it cannot select patients suited for additional adjuvant CRT. Monitoring CA19-9 levels during adjuvant therapy for postoperative patients with PDAC may guide therapeutic decisions to prevent distant failure.
Collapse
Affiliation(s)
- Yen-Feng Chiu
- Institute of Public Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Tsang-Wu Liu
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yan-Shen Shan
- Department of Surgery, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Jen-Shi Chen
- Department of Hematology-Oncology, Linkou Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Chung-Pin Li
- Divisions of Clinical Skills Training, Department of Medical Education, Taipei, Taiwan; Divisions of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ching-Liang Ho
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ruey-Kuen Hsieh
- Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Tsann-Long Hwang
- Department of Surgery, Linkou Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan; Department of Internal Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan; Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hui-Ju Ch'ang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan; Department of Radiation Oncology, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan; Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
50
|
Lee JH, Kim DK, Lee MY, Lim HS, Kwon MJ, Lee YT, Yoon KJ, Park CH. The Association of Carbohydrate Antigen (CA) 19-9 Levels and Low Skeletal Muscle Mass in Healthy Adults. Nutrients 2023; 15:3394. [PMID: 37571330 PMCID: PMC10421491 DOI: 10.3390/nu15153394] [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: 06/19/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Carbohydrate antigen 19-9 (CA 19-9) is a commonly used tumor marker for pancreatic cancer. However, CA 19-9 can be overexpressed in several benign inflammatory diseases. We investigated the relationship between high CA 19-9 level and low muscle mass (LMM) in healthy adults without cancer. Participants who underwent evaluation of muscle mass and CA 19-9 were included. Exclusion criteria were any malignancy, cardiovascular disease, tuberculosis, and chronic lung/liver disease. Participants were classified into "normal", "mild LMM", and "severe LMM" groups based on the skeletal muscle mass index. Multivariable logistic regression analyses were conducted to assess the association of high CA 19-9 with muscle mass status. A total of 263,061 adults were included. The mean age and SMI were 41.03 years and 7.13 kg/m2. After adjustments for various confounders, high CA 19-9 was independently associated with mild LMM (adjusted odds ratio, 1.677 [95% confidence interval, 1.533-1.834]) and severe LMM (2.651 [2.126-3.306]) compared to the normal group. Furthermore, the association between high CA 19-9 and severe LMM was stronger in men than in women. Elevated CA 19-9 levels were independently associated with a higher prevalence of LMM in healthy adults without cancer. Therefore, increased CA 19-9 could be utilized as a novel biomarker for sarcopenia.
Collapse
Affiliation(s)
- Jae Hyun Lee
- Department of Rehabilitation Medicine, Kosin University College of Medicine, Busan 49267, Republic of Korea;
- Department of Artificial Intelligence Convergence, Pukyong National University, Busan 48513, Republic of Korea
| | - Dong-Kun Kim
- Department of Physical and Rehabilitation Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; (D.-K.K.); (H.-S.L.); (Y.-T.L.); (K.J.Y.)
| | - Mi-Yeon Lee
- Division of Biostatistics, Department of R&D Management, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea;
| | - Han-Sol Lim
- Department of Physical and Rehabilitation Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; (D.-K.K.); (H.-S.L.); (Y.-T.L.); (K.J.Y.)
| | - Min-Jung Kwon
- Department of Laboratory Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea;
| | - Yong-Taek Lee
- Department of Physical and Rehabilitation Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; (D.-K.K.); (H.-S.L.); (Y.-T.L.); (K.J.Y.)
| | - Kyung Jae Yoon
- Department of Physical and Rehabilitation Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; (D.-K.K.); (H.-S.L.); (Y.-T.L.); (K.J.Y.)
| | - Chul-Hyun Park
- Department of Physical and Rehabilitation Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; (D.-K.K.); (H.-S.L.); (Y.-T.L.); (K.J.Y.)
| |
Collapse
|