1
|
Tang J, Li X, Tang N, Lin X, Du Y, Zhang S, Li Q, Zhang Y, Zhang Y, Hang H, Qiu T, Qiu Y, Cheng H, Dai Z, Hong H, Wei W, He J, Yan C. CD44 identified as a diagnostic biomarker for highly malignant CA19-9 negative pancreatic cancer. Cancer Lett 2025; 622:217713. [PMID: 40216152 DOI: 10.1016/j.canlet.2025.217713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/25/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with limited diagnostic biomarkers. Carbohydrate antigen 19-9 (CA19-9) is a widely used clinical biomarker and is generally considered to correlate with PDAC malignancy. However, the relationship between CA19-9 expression levels and tumor aggressiveness remains underexplored. In this study, we report a biphasic relationship between CA19-9 expression levels and PDAC malignancy, where both negative (<5 U/mL) and high (>37 U/mL) CA19-9 levels are associated with increased tumor aggressiveness. We defined CA19-9 negative PDAC as tumors that lack CA19-9 expression intracellulary, on the cell membrane, and in secreted form. In PDAC cell lines and patient-derived organoids, CA19-9 negativity, confirmed by immunofluorescence, flow cytometry and ELISA, correlated with more aggressive behaviors. In PDAC patients, tumors from those with serum CA19-9 levels below 5 U/mL exhibited stronger metabolically activity, more immunosuppressive tumor microenvironment, and worse survival than CA19-9 positive tumors, with over 90 % showing absent CA19-9 expression by immunohistochemistry (IHC). Glycoproteomics profiling identified CD44 as a highly expressed biomarker in CA19-9 negative PDAC. Elevated CD44 expression effectively distinguished CA19-9 negative PDAC from both CA19-9 positive PDAC and CA19-9 negative benign pancreatic diseases, suggesting its potential as a diagnostic tool. Furthermore, we developed a radionuclide-labeled CD44 antibody 89Zr-1M2E3, which specifically recognized CA19-9 negative PDAC tumors in preclinical models using PET-CT imaging. These findings highlight CD44 as a promising biomarker and therapeutic target for diagnosing and treating CA19-9 negative PDAC.
Collapse
Affiliation(s)
- Jiatong Tang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiaoyang Li
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Neng Tang
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiawen Lin
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yixiang Du
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China
| | - Shuo Zhang
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Qi Li
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yifan Zhang
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yixuan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China
| | - Hexing Hang
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Tongtong Qiu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yudong Qiu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Institute of Pancreatology, Nanjing University, Nanjing, Jiangsu Province, China
| | - Hao Cheng
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Zhan Dai
- Nanjing Okay Biotechnology Co., Ltd, Nanjing, Jiangsu Provinve, China
| | - Hao Hong
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Wei Wei
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China.
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Institute of Pancreatology, Nanjing University, Nanjing, Jiangsu Province, China.
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Institute of Pancreatology, Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
2
|
Liu Y, Li C, Cui X, Li M, Liu S, Wang Z. Potentially diagnostic and prognostic roles of piRNAs/PIWIs in pancreatic cancer: A review. Biochim Biophys Acta Rev Cancer 2025; 1880:189286. [PMID: 39952623 DOI: 10.1016/j.bbcan.2025.189286] [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: 12/13/2024] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with limited early diagnostic methods and therapeutic options, contributing to its poor prognosis. Recent advances in high-throughput sequencing have highlighted the critical roles of noncoding RNAs (ncRNAs), particularly PIWI-interacting RNAs (piRNAs), in cancer biology. In this review, we systematically summarize the emerging roles of piRNAs and their associated PIWI proteins in PDAC pathogenesis, progression, and prognosis. We provide a comprehensive analysis of the molecular mechanisms by which piRNAs/PIWIs regulate gene expression and cellular signaling pathways in PDAC. Furthermore, we discuss their potential as novel biomarkers for early diagnosis and therapeutic targets. Importantly, this review identifies key piRNAs/PIWIs involved in PDAC and proposes innovative strategies for improving diagnosis and treatment outcomes. Our work not only consolidates current knowledge but also offers new perspectives for future research and clinical applications in PDAC management.
Collapse
Affiliation(s)
- Yukun Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changlei Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotong Cui
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Miaomiao Li
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China
| | - Shiguo Liu
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China.
| | - Zusen Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
3
|
Dai XF, Yang YX, Yang BZ. Glycosylation editing: an innovative therapeutic opportunity in precision oncology. Mol Cell Biochem 2025; 480:1951-1967. [PMID: 38861100 DOI: 10.1007/s11010-024-05033-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 05/06/2024] [Indexed: 06/12/2024]
Abstract
Cancer is still one of the most arduous challenges in the human society, even though humans have found many ways to try to conquer it. With our incremental understandings on the impact of sugar on human health, the clinical relevance of glycosylation has attracted our attention. The fact that altered glycosylation profiles reflect and define different health statuses provide novel opportunities for cancer diagnosis and therapeutics. By reviewing the mechanisms and critical enzymes involved in protein, lipid and glycosylation, as well as current use of glycosylation for cancer diagnosis and therapeutics, we identify the pivotal connection between glycosylation and cellular redox status and, correspondingly, propose the use of redox modulatory tools such as cold atmospheric plasma (CAP) in cancer control via glycosylation editing. This paper interrogates the clinical relevance of glycosylation on cancer and has the promise to provide new ideas for laboratory practice of cold atmospheric plasma (CAP) and precision oncology therapy.
Collapse
Affiliation(s)
- Xiao-Feng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China.
| | - Yi-Xuan Yang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Bo-Zhi Yang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| |
Collapse
|
4
|
Shionoya K, Sofuni A, Mukai S, Tsuchiya T, Tanaka R, Tonozuka R, Yamamoto K, Nagai K, Matsunami Y, Kojima H, Minami H, Hirakawa N, Asano K, Yamaguchi Y, Hama K, Itoi T. Evaluating the Usefulness of the Blood Apolipoprotein A2 Isoform Index for Pancreatic Cancer Diagnosis. Cancers (Basel) 2025; 17:1071. [PMID: 40227633 PMCID: PMC11987948 DOI: 10.3390/cancers17071071] [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: 02/23/2025] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Early detection of pancreatic cancer using existing tumor markers is challenging, and novel biomarkers are needed. Apolipoprotein A2 (APOA2), which is not directly produced by tumors, may help detect pancreatic cancer through mechanisms distinct from carbohydrate antigen 19-9 (CA 19-9). This study aimed to evaluate the diagnostic performance of the APOA2-isoform (APOA2-i) Index in patients with pancreatic cancer. Methods: Serum levels of the APOA2-i Index and CA 19-9 were measured in 76 patients with pancreatic cancer (Stage 0, n = 5; I, n = 4; II, n = 15; III, n = 19; and IV, n = 33) and 98 patients with non-pancreatic cancer (intraductal papillary mucinous neoplasm, n = 36; chronic pancreatitis, n = 33; pancreatic neuroendocrine neoplasm, n = 8; autoimmune pancreatitis, n = 9; and others, n = 12) to evaluate diagnostic performance. Results: APOA2 showed lower accuracy for advanced (stages II-IV) pancreatic cancer compared to CA 19-9 (sensitivity, 50.7% vs. 83.6%; sensitivity, 77.6% vs. 87.9%), but it provided superior accuracy for early-stage (stages 0 and I) detection (sensitivity, 33.3% vs. 22.2%; specificity, 66.7% vs. 59.4%). Three early-stage pancreatic cancer cases negative for CA 19-9 were detected with the APOA2-i Index, demonstrating high diagnostic accuracy for early-stage pancreatic cancer when both biomarkers are combined (sensitivity, 44.4%; specificity, 46.7%). The multivariate analysis revealed pancreatic cancer to be an independent risk factor for APOA2-i Index positivity (odds ratio [OR]: 3.48, p < 0.001), CA 19-9 positivity (OR: 25.5, p < 0.001), and positivity for either marker (OR: 13.3, p < 0.001). Conclusions: The APOA2-i Index, combined with CA 19-9, may improve early-stage pancreatic cancer detection, especially in challenging cases and for high-risk patient surveillance.
Collapse
Affiliation(s)
- Kento Shionoya
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Atsushi Sofuni
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
- Department of Clinical Oncology, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Shuntaro Mukai
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Takayoshi Tsuchiya
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Reina Tanaka
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Ryosuke Tonozuka
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Kenjiro Yamamoto
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Kazumasa Nagai
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Yukitoshi Matsunami
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Hiroyuki Kojima
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Hirohito Minami
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Noriyuki Hirakawa
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Kyoko Asano
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Yuma Yamaguchi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Kazuki Hama
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Takao Itoi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| |
Collapse
|
5
|
Sousa P, Silva L, Câmara JS, Guedes de Pinho P, Perestrelo R. Integrating OMICS-based platforms and analytical tools for diagnosis and management of pancreatic cancer: a review. Mol Omics 2025; 21:108-121. [PMID: 39714229 DOI: 10.1039/d4mo00187g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Cancer remains the second leading cause of death worldwide, surpassed only by cardiovascular disease. From the different types of cancer, pancreatic cancer (PaC) has one of the lowest survival rates, with a survival rate of about 20% after the first year of diagnosis and about 8% after 5 years. The lack of highly sensitive and specific biomarkers, together with the absence of symptoms in the early stages, determines a late diagnosis, which is associated with a decrease in the effectiveness of medical intervention, regardless of its nature - surgery and/or chemotherapy. This review provides an updated overview of recent studies combining multi-OMICs approaches (e.g., proteomics, metabolomics) with analytical tools, highlighting the synergy between high-throughput molecular data generation and precise analytical tools such as LC-MS, GC-MS and MALDI-TOF MS. This combination significantly improves the detection, quantification and identification of biomolecules in complex biological systems and represents the latest advances in understanding PaC management and the search for effective diagnostic tools. Large-scale data analysis coupled with bioinformatics tools enables the identification of specific genetic mutations, gene expression patterns, pathways, networks, protein modifications and metabolic signatures associated with PaC pathogenesis, progression and treatment response through the integration of multi-OMICs data.
Collapse
Affiliation(s)
- Patrícia Sousa
- CQM - Centro de Química da Madeira, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal.
| | - Laurentina Silva
- Hospital Dr Nélio Mendonça, SESARAM, EPERAM - Serviço de Saúde da Região Autónoma da Madeira, Avenida Luís de Camões, 9004-514 Funchal, Portugal
| | - José S Câmara
- CQM - Centro de Química da Madeira, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal.
- Departamento de Química, Faculdade de Ciências Exatas e Engenharia, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal
| | - Paula Guedes de Pinho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, Lab. of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Rosa Perestrelo
- CQM - Centro de Química da Madeira, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal.
| |
Collapse
|
6
|
Li P, Zhang H, Chen L, Gao X, Hu Y, Xu Q, Liu W, Chen W, Chen H, Yuan S, Wang M, Liu S, Dai M. Oral and fecal microbiota as accurate non-invasive tools for detection of pancreatic cancer in the Chinese population. Cancer Lett 2025; 612:217456. [PMID: 39800212 DOI: 10.1016/j.canlet.2025.217456] [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: 11/21/2024] [Revised: 01/02/2025] [Accepted: 01/10/2025] [Indexed: 01/15/2025]
Abstract
Pancreatic cancer (PCA), a leading cause of cancer-related deaths, has limited non-invasive diagnostic methods. We aimed to identify oral and fecal microbiome biomarkers and construct diagnostic classifiers. Oral and fecal samples from 97 PCA patients and 90 healthy controls underwent 16S rRNA sequencing. Samples were randomly divided into training and validation cohorts in a 7:3 ratio. Random forest models were constructed using training cohort and validated internally and externally in Chinese, Japanese, and Spanish populations. Results revealed significant dysbiosis of the oral and fecal microbiota of PCA patients. Most of the differential taxa shared between oral and fecal samples showed similar changes. Relative abundances of Streptococcus in oral samples, and of Bifidobacterium, Klebsiella and Akkermansia in fecal samples, were enriched in PCA. The fecal Firmicutes to Bacteroidota ratio was higher in PCA patient samples. Oral and fecal microbiome classifiers based on the top 20 contributing genera were constructed, and internal validation showed that the area under the curve (AUC) values were 0.963 and 0.890, respectively. The fecal microbiome classifier performed well in the external Chinese population, with an AUC of 0.878, but poorly in the Japanese and Spanish populations. Furthermore, fecal microbiomes could predict metastasis status in PCA patients, with an AUC of 0.804. In conclusion, oral and fecal microbiota were dysbiotic in PCA patients. Fecal microbiome classifier provides a feasible, non-invasive, and cost-effective tool with high precision for PCA screening in China; oral microbiome classifier requires further validation in external populations sampled with the same simple and convenient methods.
Collapse
Affiliation(s)
- Pengyu Li
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Hanyu Zhang
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Lixin Chen
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Xingyu Gao
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Ya Hu
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Qiang Xu
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Wenjing Liu
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Weijie Chen
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Haomin Chen
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Shuai Yuan
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Mingfei Wang
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Shili Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Cheelo College of Medicine, Shandong University, No.44 Wenhuaxi Road, Lixia District, Jinan, Shandong, 250012, China
| | - Menghua Dai
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
7
|
David P, Kouhestani D, Hansen FJ, Paul S, Czubayko F, Karabiber A, Weisel N, Klösch B, Merkel S, Ole-Baur J, Gießl A, Van Deun J, Vera J, Mittelstädt A, Weber GF. Exosomal CD40, CD25, and Serum CA19-9 as Combinatory Novel Liquid Biopsy Biomarker for the Diagnosis and Prognosis of Patients with Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2025; 26:1500. [PMID: 40003965 PMCID: PMC11854914 DOI: 10.3390/ijms26041500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
The poor prognosis of pancreatic ductal adenocarcinoma (PDAC) is largely due to several challenges, such as late diagnosis, early metastasis, limited response to chemotherapy, aggressive tumor biology, and high rates of tumor recurrence. Therefore, the development of a non-invasive and effective method for early detection of PDAC is crucial to improving patient outcomes. Continued research and exploration in this area are essential to enhance early detection methods and ultimately improve the prognosis for individuals with PDAC. In this study, we examined 37 exosomal surface proteins through a multiplex flow cytometry test on peripheral plasma samples from a group of 51 clinical control individuals (including healthy volunteers and non-cancer patients (Cholecystectomy, Hernia, healthy volunteers)), 21 pancreatitis, and 48 patients diagnosed with PDAC. Our research findings revealed that the level of exosomal CD40 expression is significantly lower in patients with PDAC and pancreatitis compared to non-cancer patients (p < 0.0001). Additionally, pancreatitis patients exhibited higher levels of exosomal CD25 expression than PDAC patients (p = 0.0104). PDAC patients with higher exo-CD40 had worse survival than patients with lower exo-CD40 (p = 0.0035). Similarly, PDAC patients with higher exo-CD25 showed worse survival in comparison to patients with lower exo-CD25 (p = 0.04). Statistical analysis revealed that exosomal CD40 achieved an AUC of 0.827 in distinguishing PDAC from clinical controls. Combining exo-CD40 along with exo-CD25 and CA19-9 discriminated PDAC patients from clinical controls with an AUC of 0.92. Exo-CD40 and exo-CD25 proteins found in exosomes isolated from plasma can serve as excellent non-invasive biomarkers for the early diagnosis of PDAC. Further larger scale studies are needed to validate combined exo-CD40 and exo-CD25 as a diagnostic tool for the identification of PDAC patients through non-invasive liquid biopsy.
Collapse
Affiliation(s)
- Paul David
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (P.D.); (D.K.); (F.J.H.); (F.C.); (A.K.); (N.W.); (B.K.); (S.M.); (A.M.)
| | - Dina Kouhestani
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (P.D.); (D.K.); (F.J.H.); (F.C.); (A.K.); (N.W.); (B.K.); (S.M.); (A.M.)
| | - Frederik J. Hansen
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (P.D.); (D.K.); (F.J.H.); (F.C.); (A.K.); (N.W.); (B.K.); (S.M.); (A.M.)
| | - Sushmita Paul
- Department of Dermatology, University Hospital Erlangen, 91054 Erlangen, Germany; (S.P.); (J.O.-B.); (J.V.)
| | - Franziska Czubayko
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (P.D.); (D.K.); (F.J.H.); (F.C.); (A.K.); (N.W.); (B.K.); (S.M.); (A.M.)
| | - Alara Karabiber
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (P.D.); (D.K.); (F.J.H.); (F.C.); (A.K.); (N.W.); (B.K.); (S.M.); (A.M.)
| | - Nadine Weisel
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (P.D.); (D.K.); (F.J.H.); (F.C.); (A.K.); (N.W.); (B.K.); (S.M.); (A.M.)
| | - Bettina Klösch
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (P.D.); (D.K.); (F.J.H.); (F.C.); (A.K.); (N.W.); (B.K.); (S.M.); (A.M.)
| | - Susanne Merkel
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (P.D.); (D.K.); (F.J.H.); (F.C.); (A.K.); (N.W.); (B.K.); (S.M.); (A.M.)
| | - Jan Ole-Baur
- Department of Dermatology, University Hospital Erlangen, 91054 Erlangen, Germany; (S.P.); (J.O.-B.); (J.V.)
- Medizinische Klinik IV (Hämatologie und Onkologie), Klinikum Bayreuth GmbH, 95445 Bayreuth, Germany
| | - Andreas Gießl
- Department of Ophthalmology, University Hospital Erlangen, 91054 Erlangen, Germany;
| | - Jan Van Deun
- Department of Dermatology, University Hospital Erlangen, 91054 Erlangen, Germany; (S.P.); (J.O.-B.); (J.V.)
| | - Julio Vera
- Department of Dermatology, University Hospital Erlangen, 91054 Erlangen, Germany; (S.P.); (J.O.-B.); (J.V.)
| | - Anke Mittelstädt
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (P.D.); (D.K.); (F.J.H.); (F.C.); (A.K.); (N.W.); (B.K.); (S.M.); (A.M.)
| | - Georg F. Weber
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (P.D.); (D.K.); (F.J.H.); (F.C.); (A.K.); (N.W.); (B.K.); (S.M.); (A.M.)
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| |
Collapse
|
8
|
Cheng JJ, Matsumoto Y, Dombek GE, Stackhouse KA, Ore AS, Glickman JN, Heimburg-Molinaro J, Cummings RD. Differential expression of CD175 and CA19-9 in pancreatic adenocarcinoma. Sci Rep 2025; 15:4177. [PMID: 39905057 PMCID: PMC11794684 DOI: 10.1038/s41598-025-86988-8] [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: 05/16/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025] Open
Abstract
Alterations in protein glycosylation are observed in many solid tumor types leading to formation of tumor-associated carbohydrate antigens (TACAs). The most common TACA is the Tn antigen (CD175), which is a mucin-type O-GalNAc-Ser/Thr/Tyr glycan in membrane and secreted glycoproteins. In addition, two other TACAs are CA19-9 (sialyl-Lewis a), which is used as a prognostic serum marker for pancreatic cancer, and its isomer sialyl-Lewis x (SLex, CD15s), which is overexpressed in many cancer types and associated with metastasis. While CD175 and other TACAs may be expressed by many human carcinomas, little is known about their differential expression patterns in tumors, thus limiting their use as tissue biomarkers or therapeutic targets. Here we address the clinicopathological relevance of the expression of CA19-9, CD15s, and CD175 in pancreatic ductal adenocarcinoma (PDAC) tissues. Semi-quantitative IHC staining with well-defined monoclonal antibodies demonstrates that CD175 is expressed in all PDAC specimens analyzed. Unexpectedly, however, these TACAs are differentially expressed within PDAC specimens and their glycoproteins, but not significantly expressed in adjacent normal tissues. These data provide avenues for novel therapeutic approaches that could combine CD175- and CA19-9-targeting therapies for PDAC patients.
Collapse
Affiliation(s)
- Jane J Cheng
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, CLS-11090, Boston, MA, 02115, USA
| | - Yasuyuki Matsumoto
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, CLS-11090, Boston, MA, 02115, USA
- FDA/CDER/OBQ/OBP/DBRRIII, Silver Spring, MD, USA
| | - Gabrielle E Dombek
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, CLS-11090, Boston, MA, 02115, USA
| | - Kathryn A Stackhouse
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, CLS-11090, Boston, MA, 02115, USA
- Department of Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Ana Sofia Ore
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, CLS-11090, Boston, MA, 02115, USA
| | - Jonathan N Glickman
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, E106, Boston, MA, 02115, USA
| | - Jamie Heimburg-Molinaro
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, CLS-11090, Boston, MA, 02115, USA
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, CLS-11090, Boston, MA, 02115, USA.
| |
Collapse
|
9
|
Zardab M, Grose RP, Kocher HM. AHNAK2: a potential diagnostic biomarker for pancreatic cancer related to cellular motility. Sci Rep 2025; 15:2934. [PMID: 39849106 PMCID: PMC11757713 DOI: 10.1038/s41598-025-87337-5] [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: 09/16/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025] Open
Abstract
Pancreatic ductal adenocarcinoma lacks suitable biomarkers for early diagnosis of disease. In gene panels developed for early diagnosis of pancreatic cancer, high AHNAK2 mRNA expression was one possible biomarker. In silico analysis of published human sample datasets (n = 177) and ex vivo analysis of human plasma samples (n = 30 PDAC with matched 30 healthy control) suggested AHNAK2 could be a diagnostic biomarker. At a plasma level of 421.47 ng/ml, AHNAK2 could potentially diagnose PDAC with a specificity and sensitivity of 83.33% and 86.67%. In vitro analysis suggests that in cell lines with diffuse cytoplasmic distribution of AHNAK2, there was colocalization of AHNAK2 with Cortactin in filipodia. This colocalization increased when cells were cultured on substrates such as Fibronectin and Collagen, as well as in hypoxia, and resulted in an augmented invasion of cancer cells. However, in cell lines with a vesicular AHNAK2 staining, such changes were not observed. Our study posits AHNAK2 as a valuable diagnostic biomarker in PDAC, now demanding prospective validation. Determination of mechanisms regulating AHNAK2 subcellular localisation may help explain its biological role.
Collapse
MESH Headings
- Humans
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/blood
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
- Cell Movement
- Cell Line, Tumor
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/blood
- Carcinoma, Pancreatic Ductal/metabolism
- Male
- Female
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/blood
- Cytoskeletal Proteins/metabolism
- Middle Aged
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neoplasm Proteins/blood
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Cortactin/metabolism
- Gene Expression Regulation, Neoplastic
- Aged
Collapse
Affiliation(s)
- Mohamed Zardab
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Hemant M Kocher
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
- Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, London, UK.
| |
Collapse
|
10
|
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
|
11
|
Yu CW, Wu LW, Hu JM, Chang PK. Serum CA19-9 as a predictor of incident metabolic syndrome in obese middle-aged and older men: A 9-year cohort study. Cancer Biomark 2025; 42:18758592241296282. [PMID: 40109214 DOI: 10.1177/18758592241296282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
BackgroundThe status of carbohydrate antigen 19-9 (CA19-9) in metabolic syndrome (MetS) is unknown.ObjectiveTo investigate the association between serum CA19-9 levels and incident metabolic syndrome in obese middle-aged and older men.MethodsFrom 2007 to 2015, 1,750 participants were retrospectively reviewed. Health checkup data were obtained, and participants were divided into three groups based on CA19-9 levels. Various parameters including BMI, waist circumference, blood pressure, and biochemical parameters were measured. Cox regression analysis was used to assess the association between CA19-9 levels and incident MetS. The MetS diagnostic criteria were based on the National Cholesterol Education Program Adult Treatment Panel III guidelines.ResultsThe highest CA19-9 tertile was associated with an increased risk of incident MetS, high systolic blood pressure, high waist circumference, high fasting plasma glucose, low high-density lipoprotein, and high triglyceride levels. The observation period was 9 years, during which 328 (18.7%) new-onset MetS cases were identified. Subgroup analysis showed increased risk among individuals in the highest CA19-9 tertile who were obese, male, and ≥ 50 years old.ConclusionsThere is a positive correlation between serum CA19-9 levels and incident metabolic syndrome in obese middle-aged and older men.
Collapse
Affiliation(s)
- Chun-Wei Yu
- Division of Colon and Rectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Republic of China
| | - Li-Wei Wu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Republic of China
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Republic of China
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Republic of China
| | - Je-Ming Hu
- Division of Colon and Rectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Republic of China
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Republic of China
| | - Pi-Kai Chang
- Division of Colon and Rectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Republic of China
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Republic of China
| |
Collapse
|
12
|
Tajbakhsh J, Debernardi S, Blyuss O, Bai J, Weng R, Lo S, Pandol SJ, Crnogorac-Jurcevic T, Gupta NK. A Clinical Laboratory Improvement Amendments/College of American Pathologists-Compliant Noninvasive Laboratory-Developed Test for Early Detection of Pancreatic Ductal Adenocarcinoma. J Mol Diagn 2025; 27:54-61. [PMID: 39521243 DOI: 10.1016/j.jmoldx.2024.10.001] [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: 12/11/2023] [Revised: 09/01/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
A noninvasive test for earlier detection of pancreatic cancer in individuals at higher risk is currently unavailable. We devised PancSure, a laboratory-developed test based on the protein biomarkers lymphatic vessel endothelial hyaluronan receptor 1 (LYVE1) and regenerating family member 1 β (REG1B), measured in urine by enzyme-linked immunosorbent assay, and commonly used serum/plasma carbohydrate antigen 19.9 (CA19.9), with an updated PancRISK algorithm for data interpretation. The test was validated in 565 patients: 117 asymptomatic patients without any known pancreatic condition or malignancies (21%), 242 symptomatic patients with benign pancreatic diseases (43%), and 206 patients with confirmed cancers (36%); 161 (77.5%) had stage I to II disease, and 45 (22.5%) had stage III to IV disease. PancSure passed all specifications during analytical validation and distinguishes early-stage resectable cancer from asymptomatic individuals with area under the receiver operating characteristic curve (AUC) of 0.93 (95% CI, 0.89-0.97) and 85% to 90% sensitivity (SN) and 78% to 87% specificity (SP); from symptomatic patients with AUC of 0.86 (95% CI, 0.81-0.91) and 83% to 85% SN and 72% to 83% SP; and from all noncancer patients (pooled controls) with AUC of 0.89 (95% CI, 0.84-0.93) and 83% to 85% SN and 78% to 87% SP. PancSure is a noninvasive clinical-grade test with a 48-hour turnover, ready for implementation, providing a viable solution for the earlier detection of pancreatic cancer in at-risk groups for improved patient care.
Collapse
Affiliation(s)
- Jian Tajbakhsh
- 3rd Street Diagnostics, Cedars-Sinai, Los Angeles, California; The Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai, Los Angeles, California.
| | - Silvana Debernardi
- The Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Oleg Blyuss
- The Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - Jianhao Bai
- The Diagnostic Development Hub, Agency for Science, Technology and Research, Singapore, Singapore
| | - Ruifen Weng
- The Diagnostic Development Hub, Agency for Science, Technology and Research, Singapore, Singapore
| | - Simon Lo
- Department of Medicine, Cedars-Sinai, Los Angeles, California
| | | | | | - Nirdesh K Gupta
- 3rd Street Diagnostics, Cedars-Sinai, Los Angeles, California
| |
Collapse
|
13
|
Shan H, Tian G, Zhang Y, Qiu Z. Exploring the molecular mechanisms and therapeutic potential of SMAD4 in colorectal cancer. Cancer Biol Ther 2024; 25:2392341. [PMID: 39164192 PMCID: PMC11340766 DOI: 10.1080/15384047.2024.2392341] [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: 05/21/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024] Open
Abstract
Colorectal Cancer (CRC) is the third most common cancer worldwide, and the occurrence and development of CRC are influenced by the molecular biology characteristics of CRC, especially alterations in key signaling pathways. The transforming growth factor-β (TGF-β) plays a crucial role in cellular growth, differentiation, migration, and apoptosis, with SMAD4 protein serving as a key transcription factor in the TGF-β signaling pathway, thus playing a significant role in the onset and progression of CRC. CRC is one of the malignancies with a high mortality rate worldwide. Despite significant research progress in recent years, especially regarding the role of SMAD4, its dual role in the early and late stages of tumor progression has promoted further discussion on its complexity as a therapeutic target, highlighting the urgent need for a deeper analysis of its role in CRC. This review aims to explore the function of SMAD4 protein in CRC and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Hui Shan
- Department of Oncology, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Guangyu Tian
- Department of Oncology, Jiangdu People’s Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Yeqing Zhang
- Department of Vascular Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhiyuan Qiu
- Department of Oncology, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
14
|
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
|
15
|
Vitale F, Zileri Dal Verme L, Paratore M, Negri M, Nista EC, Ainora ME, Esposto G, Mignini I, Borriello R, Galasso L, Alfieri S, Gasbarrini A, Zocco MA, Nicoletti A. The Past, Present, and Future of Biomarkers for the Early Diagnosis of Pancreatic Cancer. Biomedicines 2024; 12:2840. [PMID: 39767746 PMCID: PMC11673965 DOI: 10.3390/biomedicines12122840] [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: 11/01/2024] [Revised: 11/30/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
Pancreatic cancer is one of the most aggressive cancers with a very poor 5-year survival rate and reduced therapeutic options when diagnosed in an advanced stage. The dismal prognosis of pancreatic cancer has guided significant efforts to discover novel biomarkers in order to anticipate diagnosis, increasing the population of patients who can benefit from curative surgical treatment. CA 19-9 is the reference biomarker that supports the diagnosis and guides the response to treatments. However, it has significant limitations, a low specificity, and is inefficient as a screening tool. Several potential biomarkers have been discovered in the serum, urine, feces, and pancreatic juice of patients. However, most of this evidence needs further validation in larger cohorts. The advent of advanced omics sciences and liquid biopsy techniques has further enhanced this field of research. The aim of this review is to analyze the historical evolution of the research on novel biomarkers for the early diagnosis of pancreatic cancer, focusing on the current evidence for the most promising biomarkers from different body fluids and the novel trends in research, such as omics sciences and liquid biopsy, in order to favor the application of modern personalized medicine.
Collapse
Affiliation(s)
- Federica Vitale
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Lorenzo Zileri Dal Verme
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Mattia Paratore
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Marcantonio Negri
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Enrico Celestino Nista
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Maria Elena Ainora
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Giorgio Esposto
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Irene Mignini
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Raffaele Borriello
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Linda Galasso
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Sergio Alfieri
- Centro Pancreas, Chirurgia Digestiva, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Maria Assunta Zocco
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Alberto Nicoletti
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| |
Collapse
|
16
|
Nakamura Y, Yasukawa T, Fukumura Y, Takeda Y, Imamura H, Shi Y, Li M, Abe M, Uyama S, Kajino K, Ishijima M, Saiura A, Orimo A. Association of stromal type IV collagen and prognosis in neoadjuvant chemotherapy-treated pancreatic cancer. Jpn J Clin Oncol 2024; 54:1261-1271. [PMID: 39180719 PMCID: PMC11631120 DOI: 10.1093/jjco/hyae118] [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: 05/31/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has poor prognosis due to its low surgical eligibility and resistance to chemotherapy. Abundant stroma is characteristic of PDAC, and cancer-associated fibroblasts (CAFs) are a major stromal constituent, contributing to chemoresistance. Because neoadjuvant chemotherapy (NAC) is included in PDAC treatment as a standard regimen, the role of CAFs in NAC resistance must be studied. Although type IV collagen (COLIV) is present in the tumor of PDAC, the association between COLIV and disease advancement of NAC-treated PDAC is unclear. METHODS Using a cohort of NAC-treated patients with PDAC, we examined clinicopathological data and conducted immunohistochemical analysis of COLIV in tissue specimens prepared from surgically resected pancreas. RESULTS AND CONCLUSIONS Our analysis revealed that ~50% of the cases were positive for COLIV in the stroma and diffuse COLIV staining was an independent poor prognosis factor alongside high serum CA19-9 before NAC treatment (>37 U/mL) and postsurgical residual tumors. Based on these findings, we propose that stromal COLIV staining can be used to predict prognosis in NAC-treated patients with PDAC after surgery. Additionally, these findings suggest a possibility that stromal COLIV staining indicates resistance to anticancer drugs and/or contributes to malignancy in PDAC.
Collapse
Affiliation(s)
- Yasuhiro Nakamura
- Department of Orthopaedics, Faculty of Medicine, Juntendo University, Tokyo, Japan
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Molecular Pathogenesis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takehiro Yasukawa
- Department of Molecular Pathogenesis, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Pathology and Oncology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yuki Fukumura
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoshinori Takeda
- Department of Hepatobiliary and Pancreatic Surgery, Juntendo University School of Medicine, Tokyo Japan
| | - Hiroshi Imamura
- Department of Hepatobiliary and Pancreatic Surgery, Juntendo University School of Medicine, Tokyo Japan
| | - Yang Shi
- Department of Molecular Pathogenesis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mu Li
- Department of Molecular Pathogenesis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masaaki Abe
- Department of Pathology and Oncology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Saya Uyama
- Department of Pathology and Oncology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Kazunori Kajino
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Muneaki Ishijima
- Department of Orthopaedics, Faculty of Medicine, Juntendo University, Tokyo, Japan
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akio Saiura
- Department of Hepatobiliary and Pancreatic Surgery, Juntendo University School of Medicine, Tokyo Japan
| | - Akira Orimo
- Department of Molecular Pathogenesis, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Pathology and Oncology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
17
|
Zhang W, Zhao C, Jia H, Liu T, Yang J, Wu P, Mu X. Biomarker detection based on nanoparticle-induced ultrasonic Rayleigh scattering. MICROSYSTEMS & NANOENGINEERING 2024; 10:182. [PMID: 39632870 PMCID: PMC11618333 DOI: 10.1038/s41378-024-00808-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/02/2024] [Accepted: 09/20/2024] [Indexed: 12/07/2024]
Abstract
Ultrasonic biochemical detection is important for biomarker detection, drug monitoring, and medical diagnosis, as it can predict disease progression and enable effective measures to be taken in a timely manner. However, the ultrasonic technology currently used for biochemical marker detection is directly modified on the surface of the device. The associated test methods are costly and unreliable while having poor repeatability; therefore, they cannot achieve low-cost rapid testing. In this study, a detection mechanism based on the Rayleigh scattering of acoustic waves caused by nanoparticles, which causes changes in the received sound pressure, was developed for the first time. The modification of antibodies on an insertable substrate decouples the functionalization step from the sensor surface and facilitates the application of capacitive micromachined ultrasonic transducers (CMUTs) in conjunction with Au nanoparticles (AuNPs) for CA19-9 cancer antigen detection. A corresponding detection theory was established, and the relevant parameters of the theoretical formula were verified using different nanoparticles. Using our fabricated CMUT chip with a resonant frequency of 1 MHz, the concentrations and substances of the CA19-9 antigen markers were successfully measured. In the concentration range of 0.1-1000 U/mL, the receiving voltage decreased with increasing concentration. Further investigations revealed that the influence of other interfering markers in the human body can be ignored, demonstrating the feasibility and robustness of biochemical detection based on CMUTs combined with nanoparticles.
Collapse
Affiliation(s)
- Wangyang Zhang
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education and International Research and Development Center of Micro-Nano Systems and New Materials Technology, Chongqing University, Chongqing, 400044, China
| | - Chaoshan Zhao
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education and International Research and Development Center of Micro-Nano Systems and New Materials Technology, Chongqing University, Chongqing, 400044, China
| | - Haoliang Jia
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education and International Research and Development Center of Micro-Nano Systems and New Materials Technology, Chongqing University, Chongqing, 400044, China
| | - Tao Liu
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education and International Research and Development Center of Micro-Nano Systems and New Materials Technology, Chongqing University, Chongqing, 400044, China
| | - Jiaqian Yang
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education and International Research and Development Center of Micro-Nano Systems and New Materials Technology, Chongqing University, Chongqing, 400044, China
| | - Pengfan Wu
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education and International Research and Development Center of Micro-Nano Systems and New Materials Technology, Chongqing University, Chongqing, 400044, China
| | - Xiaojing Mu
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education and International Research and Development Center of Micro-Nano Systems and New Materials Technology, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
18
|
Su N, Tang R, Zhang Y, Ni J, Huang Y, Liu C, Xiao Y, Zhu B, Zhao Y. Using machine learning to identify risk factors for pancreatic cancer: a retrospective cohort study of real-world data. Front Pharmacol 2024; 15:1510220. [PMID: 39640479 PMCID: PMC11617206 DOI: 10.3389/fphar.2024.1510220] [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: 10/12/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024] Open
Abstract
Objectives This study aimed to identify the risk factors for pancreatic cancer through machine learning. Methods We investigated the relationships between different risk factors and pancreatic cancer using a real-world retrospective cohort study conducted at West China Hospital of Sichuan University. Multivariable logistic regression, with pancreatic cancer as the outcome, was used to identify covariates associated with pancreatic cancer. The machine learning model extreme gradient boosting (XGBoost) was adopted as the final model for its high performance. Shapley additive explanations (SHAPs) were utilized to visualize the relationships between these potential risk factors and pancreatic cancer. Results The cohort included 1,982 patients. The median ages for pancreatic cancer and nonpancreatic cancer groups were 58.1 years (IQR: 51.3-64.4) and 57.5 years (IQR: 49.5-64.9), respectively. Multivariable logistic regression indicated that kirsten rats arcomaviral oncogene homolog (KRAS) gene mutation, hyperlipidaemia, pancreatitis, and pancreatic cysts are significantly correlated with an increased risk of pancreatic cancer. The five most highly ranked features in the XGBoost model were KRAS gene mutation status, age, alcohol consumption status, pancreatitis status, and hyperlipidaemia status. Conclusion Machine learning algorithms confirmed that KRAS gene mutation, hyperlipidaemia, and pancreatitis are potential risk factors for pancreatic cancer. Additionally, the coexistence of KRAS gene mutation and pancreatitis, as well as KRAS gene mutation and pancreatic cysts, is associated with an increased risk of pancreatic cancer. Our findings offered valuable implications for public health strategies targeting the prevention and early detection of pancreatic cancer.
Collapse
Affiliation(s)
- Na Su
- West China School of Pharmacy, Sichuan University, Chengdu, China
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Tang
- Institute of Medical Information, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Yice Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jiaqi Ni
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yimei Huang
- University of Florida Health Shands Hospital, Gainesville, FL, United States
| | - Chunqi Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuzhou Xiao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Baoting Zhu
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Qiu X, Liu W, Xing XY, Chen LJ. A case of extremely high serum CA19-9 level without any inducement. Asian J Surg 2024:S1015-9584(24)02454-0. [PMID: 39528371 DOI: 10.1016/j.asjsur.2024.10.129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Affiliation(s)
- Xuan Qiu
- Department of Faculty Surgery, St. Petersburg State University, Saint Petersburg, Russia; Department of General Surgery, Shandong Linglong Yingcheng Hospital, Zhaoyuan, Yantai, China
| | - Wei Liu
- Department of Oncology, Qingdao Hiser Hospital Affiliated of Qingdao University, Tsingtao, China
| | - Xiao-Yan Xing
- Department of Oncology, Qingdao Hiser Hospital Affiliated of Qingdao University, Tsingtao, China
| | - Li-Juan Chen
- Department of Oncology, Qingdao Hiser Hospital Affiliated of Qingdao University, Tsingtao, China.
| |
Collapse
|
20
|
Wang T, Bao X, Yang F, Pan S, Xu K, Ren T. Plasma COL10A1 Level, a Potential Diagnostic and Prognostic Biomarker for Pancreatic Ductal Adenocarcinoma. Onco Targets Ther 2024; 17:949-959. [PMID: 39525356 PMCID: PMC11550710 DOI: 10.2147/ott.s474540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND COL10A1 expression was up-regulated and could promote tumor development in pancreatic cancer. As a secreted protein, plasma COL10A1 level was proven to have certain diagnostic efficacy in gastric cancer, breast cancer, and colorectal cancer. It is still unknown whether it has a biomarker role for pancreatic cancer. AIM To explore and analyze the diagnostic and prognostic value of plasma COL10A1 level in pancreatic ductal adenocarcinoma (PDAC). METHOD The RNA-seq dataset of PDAC from The Cancer Genome Atlas (TCGA) and six expression profiling microarray datasets from Gene Expression Omnibus (GEO) were downloaded to analyze the expression of COL10A1 in tissues. Thirty-six patients with PDAC and eighteen healthy volunteers were enrolled to measure COL10A1 levels in tissues and plasmas, and the relationship between clinical characteristics and the COL10A1 levels was analyzed. The diagnostic and prognostic efficacy of plasma COL10A1 levels were calculated. RESULTS Aspects of COL10A1 expression level in tissues, COL10A1 expression was significantly higher in PDAC tissue than adjacent normal tissue. The expression of COL10A1 was correlated with T, M, and AJCC stages. Patients with high COL10A1 expression had worse recurrence-free survival (RFS) and overall survival (OS) than those with low expression. Aspects of COL10A1 expression levels in plasma, its diagnostic area under the curve (AUC) for PDAC was 0.926 (95% CI 0.853-0.999), diagnostic sensitivity was 81% (95% CI 64-92%), and specificity was 100% (95% CI 81-100%). The time-dependent AUCs at 1-year and 3-year were 0.71 (95% CI 0.51-0.90) and 0.74 (95% CI 0.48-1.00), respectively. CONCLUSION In PDAC, plasma COL10A1 levels showed certain diagnostic and prognostic efficacy. COL10A1 may be a diagnostic and prognostic biomarker for PDAC and play a role in liquid biopsy of this disease.
Collapse
Affiliation(s)
- Tianlei Wang
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, People’s Republic of China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People’s Republic of China
| | - Xinrui Bao
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, People’s Republic of China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People’s Republic of China
| | - Fang Yang
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, People’s Republic of China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People’s Republic of China
| | - Shenbin Pan
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, People’s Republic of China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People’s Republic of China
| | - Ke Xu
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, People’s Republic of China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People’s Republic of China
| | - Tao Ren
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, People’s Republic of China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
21
|
Haab B, Qian L, Staal B, Jain M, Fahrmann J, Worthington C, Prosser D, Velokokhatnaya L, Lopez C, Tang R, Hurd MW, Natarajan G, Kumar S, Smith L, Hanash S, Batra SK, Maitra A, Lokshin A, Huang Y, Brand RE. A rigorous multi-laboratory study of known PDAC biomarkers identifies increased sensitivity and specificity over CA19-9 alone. Cancer Lett 2024; 604:217245. [PMID: 39276915 PMCID: PMC11808537 DOI: 10.1016/j.canlet.2024.217245] [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: 05/10/2024] [Revised: 08/24/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
A blood test that enables surveillance for early-stage pancreatic ductal adenocarcinoma (PDAC) is an urgent need. Independent laboratories have reported PDAC biomarkers that could improve biomarker performance over CA19-9 alone, but the performance of the previously reported biomarkers in combination is not known. Therefore, we conducted a coordinated case/control study across multiple laboratories using common sets of blinded training and validation samples (132 and 295 plasma samples, respectively) from PDAC patients and non-PDAC control subjects representing conditions under which surveillance occurs. We analyzed the training set to identify candidate biomarker combination panels using biomarkers across laboratories, and we applied the fixed panels to the validation set. The panels identified in the training set, CA19-9 with CA199.STRA, LRG1, TIMP-1, TGM2, THSP2, ANG, and MUC16.STRA, achieved consistent performance in the validation set. The panel of CA19-9 with the glycan biomarker CA199.STRA improved sensitivity from 0.44 with 0.98 specificity for CA19-9 alone to 0.71 with 0.98 specificity (p < 0.001, 1000-fold bootstrap). Similarly, CA19-9 combined with the protein biomarker LRG1 and CA199.STRA improved specificity from 0.16 with 0.94 sensitivity for CA19-9 to 0.65 with 0.89 sensitivity (p < 0.001, 1000-fold bootstrap). We further validated significantly improved performance using biomarker panels that did not include CA19-9. This study establishes the effectiveness of a coordinated study of previously discovered biomarkers and identified panels of those biomarkers that significantly increased the sensitivity and specificity of early-stage PDAC detection in a rigorous validation trial.
Collapse
Affiliation(s)
- Brian Haab
- Van Andel Institute, 333 Bostwick NE, Grand Rapids, MI, 49503, USA.
| | - Lu Qian
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 19024, USA
| | - Ben Staal
- Van Andel Institute, 333 Bostwick NE, Grand Rapids, MI, 49503, USA
| | - Maneesh Jain
- University of Nebraska Medical Center, 42nd and Emile Streets, Omaha, NE, 68198, USA
| | - Johannes Fahrmann
- MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Christine Worthington
- University of Pittsburgh Medical Center, 200 Lothrop St., Pittsburgh, PA, 15213-2582, USA
| | - Denise Prosser
- University of Pittsburgh Medical Center, 200 Lothrop St., Pittsburgh, PA, 15213-2582, USA
| | | | - Camden Lopez
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 19024, USA
| | - Runlong Tang
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 19024, USA
| | - Mark W Hurd
- MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | | | - Sushil Kumar
- University of Nebraska Medical Center, 42nd and Emile Streets, Omaha, NE, 68198, USA
| | - Lynette Smith
- University of Nebraska Medical Center, 42nd and Emile Streets, Omaha, NE, 68198, USA
| | - Sam Hanash
- MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Surinder K Batra
- University of Nebraska Medical Center, 42nd and Emile Streets, Omaha, NE, 68198, USA
| | - Anirban Maitra
- MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Anna Lokshin
- University of Pittsburgh Medical Center, 200 Lothrop St., Pittsburgh, PA, 15213-2582, USA
| | - Ying Huang
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 19024, USA
| | - Randall E Brand
- University of Pittsburgh Medical Center, 200 Lothrop St., Pittsburgh, PA, 15213-2582, USA.
| |
Collapse
|
22
|
Jin M, Liu HL, Xue J, Ma H, Liu JL, Lin ZY, Wang J, Bao LQ, Luo ZG, Yu XJ, Li S, Hu JL, Zhang T. Nab-paclitaxel plus S-1 versus nab-paclitaxel plus gemcitabine in patients with advanced pancreatic cancer: a multicenter, randomized, phase II study. Oncologist 2024; 29:e1406-e1418. [PMID: 38990195 PMCID: PMC11449102 DOI: 10.1093/oncolo/oyae171] [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: 02/28/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Encouraging antitumor activity of nab-paclitaxel plus S-1 (AS) has been shown in several small-scale studies. This study compared the efficacy and safety of AS versus standard-of-care nab-paclitaxel plus gemcitabine (AG) as a first-line treatment for advanced pancreatic cancer (PC). METHODS In this multicenter, randomized, phase II trial, eligible patients with unresectable, locally advanced, or metastatic PC were recruited and randomly assigned (1:1) to receive AS (nab-paclitaxel 125 mg/m2 on days 1 and 8; S-1 twice daily on days 1 through 14) or AG (nab-paclitaxel 125 mg/m2 on days 1 and 8; gemcitabine 1000 mg/m2 on days 1 and 8) for 6 cycles. The primary endpoint was progression-free survival (PFS). RESULTS Between July 16, 2019, and September 9, 2022, 62 patients (AS, n = 32; AG, n = 30) were treated and evaluated. With a median follow-up of 8.36 months at preplanned interim analysis (data cutoff, March 24, 2023), the median PFS (8.48 vs 4.47 months; hazard ratio [HR], 0.402; P = .002) and overall survival (OS; 13.73 vs 9.59 months; HR, 0.226; P < .001) in the AS group were significantly longer compared to the AG group. More patients had objective response in the AS group than AG group (37.50% vs 6.67%; P = .005). The most common grade 3-4 adverse events were neutropenia and leucopenia in both groups, and gamma glutamyl transferase increase was observed only in the AG group. CONCLUSION The first-line AS regimen significantly extended both PFS and OS of Chinese patients with advanced PC when compared with the AG regimen, with a comparable safety profile. (ClinicalTrials.gov Identifier: NCT03636308).
Collapse
Affiliation(s)
- Min Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, People’s Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Hong-Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, People’s Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Jun Xue
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, People’s Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Hong Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, People’s Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Jun-Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, People’s Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Zhen-Yu Lin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, People’s Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Jing Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, People’s Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Le-Qun Bao
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Zhi-Guo Luo
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, People’s Republic of China
| | - Xiong-Jie Yu
- Department of Oncology, Shiyan People’s Hospital, Shiyan, 442000, People’s Republic of China
| | - Shuang Li
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, 434000, People’s Republic of China
| | - Jian-Li Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, People’s Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, People’s Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| |
Collapse
|
23
|
Daurai B, Baruah AJ, Gogoi M. Recent advances in point-of-care biosensors for pancreatic diseases. Trends Analyt Chem 2024; 179:117867. [DOI: 10.1016/j.trac.2024.117867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
24
|
Borgmästars E, Ulfenborg B, Johansson M, Jonsson P, Billing O, Franklin O, Lundin C, Jacobson S, Simm M, Lubovac-Pilav Z, Sund M. Multi-omics profiling to identify early plasma biomarkers in pre-diagnostic pancreatic ductal adenocarcinoma: a nested case-control study. Transl Oncol 2024; 48:102059. [PMID: 39018772 PMCID: PMC11301391 DOI: 10.1016/j.tranon.2024.102059] [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: 04/05/2024] [Revised: 05/20/2024] [Accepted: 07/05/2024] [Indexed: 07/19/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with poor survival. Novel biomarkers are urgently needed to improve the outcome through early detection. Here, we aimed to discover novel biomarkers for early PDAC detection using multi-omics profiling in pre-diagnostic plasma samples biobanked after routine health examinations. A nested case-control study within the Northern Sweden Health and Disease Study was designed. Pre-diagnostic plasma samples from 37 future PDAC patients collected within 2.3 years before diagnosis and 37 matched healthy controls were included. We analyzed metabolites using liquid chromatography mass spectrometry and gas chromatography mass spectrometry, microRNAs by HTG edgeseq, proteins by multiplex proximity extension assays, as well as three clinical biomarkers using milliplex technology. Supervised and unsupervised multi-omics integration were performed as well as univariate analyses for the different omics types and clinical biomarkers. Multiple hypothesis testing was corrected using Benjamini-Hochberg's method and a false discovery rate (FDR) below 0.1 was considered statistically significant. Carbohydrate antigen (CA) 19-9 was associated with PDAC risk (OR [95 % CI] = 3.09 [1.31-7.29], FDR = 0.03) and increased closer to PDAC diagnosis. Supervised multi-omics models resulted in poor discrimination between future PDAC cases and healthy controls with obtained accuracies between 0.429-0.500. No single metabolite, microRNA, or protein was differentially altered (FDR < 0.1) between future PDAC cases and healthy controls. CA 19-9 levels increase up to two years prior to PDAC diagnosis but extensive multi-omics analysis including metabolomics, microRNAomics and proteomics in this cohort did not identify novel early biomarkers for PDAC.
Collapse
Affiliation(s)
- Emmy Borgmästars
- Department of Diagnostics and Intervention/ Surgery, Umeå University, Umeå, Sweden.
| | - Benjamin Ulfenborg
- School of Bioscience, Department of Biology and Bioinformatics, University of Skövde, Skövde, Sweden
| | - Mattias Johansson
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Pär Jonsson
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Ola Billing
- Department of Diagnostics and Intervention/ Surgery, Umeå University, Umeå, Sweden
| | - Oskar Franklin
- Department of Diagnostics and Intervention/ Surgery, Umeå University, Umeå, Sweden; Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, USA
| | - Christina Lundin
- Department of Diagnostics and Intervention/ Surgery, Umeå University, Umeå, Sweden
| | - Sara Jacobson
- Department of Diagnostics and Intervention/ Surgery, Umeå University, Umeå, Sweden
| | - Maja Simm
- Department of Diagnostics and Intervention/ Surgery, Umeå University, Umeå, Sweden; Department of Clinical Sciences/ Obstetrics and Gynecology, Umeå University, Umeå, Sweden
| | - Zelmina Lubovac-Pilav
- School of Bioscience, Department of Biology and Bioinformatics, University of Skövde, Skövde, Sweden
| | - Malin Sund
- Department of Diagnostics and Intervention/ Surgery, Umeå University, Umeå, Sweden; Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
25
|
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
|
26
|
Fuller RN, Morcos A, Bustillos JG, Molina DC, Wall NR. Small non-coding RNAs and pancreatic ductal adenocarcinoma: Linking diagnosis, pathogenesis, drug resistance, and therapeutic potential. Biochim Biophys Acta Rev Cancer 2024; 1879:189153. [PMID: 38986720 DOI: 10.1016/j.bbcan.2024.189153] [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: 01/22/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024]
Abstract
This review comprehensively investigates the intricate interplay between small non-coding RNAs (sncRNAs) and pancreatic ductal adenocarcinoma (PDAC), a devastating malignancy with limited therapeutic options. Our analysis reveals the pivotal roles of sncRNAs in various facets of PDAC biology, spanning diagnosis, pathogenesis, drug resistance, and therapeutic strategies. sncRNAs have emerged as promising biomarkers for PDAC, demonstrating distinct expression profiles in diseased tissues. sncRNA differential expression patterns, often detectable in bodily fluids, hold potential for early and minimally invasive diagnostic approaches. Furthermore, sncRNAs exhibit intricate involvement in PDAC pathogenesis, regulating critical cellular processes such as proliferation, apoptosis, and metastasis. Additionally, mechanistic insights into sncRNA-mediated pathogenic pathways illuminate novel therapeutic targets and interventions. A significant focus of this review is dedicated to unraveling sncRNA mechanisms underlying drug resistance in PDAC. Understanding these mechanisms at the molecular level is imperative for devising strategies to overcome drug resistance. Exploring the therapeutic landscape, we discuss the potential of sncRNAs as therapeutic agents themselves as their ability to modulate gene expression with high specificity renders them attractive candidates for targeted therapy. In summary, this review integrates current knowledge on sncRNAs in PDAC, offering a holistic perspective on their diagnostic, pathogenic, and therapeutic relevance. By elucidating the roles of sncRNAs in PDAC biology, this review provides valuable insights for the development of novel diagnostic tools and targeted therapeutic approaches, crucial for improving the prognosis of PDAC patients.
Collapse
Affiliation(s)
- Ryan N Fuller
- Department of Basic Science, Division of Biochemistry, Center for Health Disparity and Mol. Med., Loma Linda University, Loma Linda, CA 92350, USA; Department of Radiation Medicine, James M. Slater, MD Proton Treatment and Research Center, Loma Linda University, Loma Linda, CA 92350, USA
| | - Ann Morcos
- Department of Basic Science, Division of Biochemistry, Center for Health Disparity and Mol. Med., Loma Linda University, Loma Linda, CA 92350, USA; Department of Radiation Medicine, James M. Slater, MD Proton Treatment and Research Center, Loma Linda University, Loma Linda, CA 92350, USA
| | - Joab Galvan Bustillos
- Department of Basic Science, Division of Biochemistry, Center for Health Disparity and Mol. Med., Loma Linda University, Loma Linda, CA 92350, USA; Division of Surgical Oncology, Department of Surgery, Loma Linda University, Loma Linda, CA 92350, USA
| | - David Caba Molina
- Division of Surgical Oncology, Department of Surgery, Loma Linda University, Loma Linda, CA 92350, USA
| | - Nathan R Wall
- Department of Basic Science, Division of Biochemistry, Center for Health Disparity and Mol. Med., Loma Linda University, Loma Linda, CA 92350, USA; Department of Radiation Medicine, James M. Slater, MD Proton Treatment and Research Center, Loma Linda University, Loma Linda, CA 92350, USA.
| |
Collapse
|
27
|
Bugazia D, Al-Najjar E, Esmail A, Abdelrahim S, Abboud K, Abdelrahim A, Umoru G, Rayyan HA, Abudayyeh A, Al Moustafa AE, Abdelrahim M. Pancreatic ductal adenocarcinoma: the latest on diagnosis, molecular profiling, and systemic treatments. Front Oncol 2024; 14:1386699. [PMID: 39011469 PMCID: PMC11247645 DOI: 10.3389/fonc.2024.1386699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/30/2024] [Indexed: 07/17/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is currently the fourth leading cause of death in the United States and is expected to be ranked second in the next 10 years due to poor prognosis and a rising incidence. Distant metastatic PDAC is associated with the worst prognosis among the different phases of PDAC. The diagnostic options for PDAC are convenient and available for staging, tumor response evaluation, and management of resectable or borderline resectable PDAC. However, imaging is crucial in PDAC diagnosis, monitoring, resectability appraisal, and response evaluation. The advancement of medical technologies is evolving, hence the use of imaging in PDAC treatment options has grown as well as the utilization of ctDNA as a tumor marker. Treatment options for metastatic PDAC are minimal with the primary goal of therapy limited to symptom relief or palliation, especially in patients with low functional capacity at the point of diagnosis. Molecular profiling has shown promising potential solutions that would push the treatment boundaries for patients with PDAC. In this review, we will discuss the latest updates from evidence-based guidelines regarding diagnosis, therapy response evaluation, prognosis, and surveillance, as well as illustrating novel therapies that have been recently investigated for PDAC, in addition to discussing the molecular profiling advances in PDAC.
Collapse
Affiliation(s)
- Doaa Bugazia
- Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Ebtesam Al-Najjar
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX, United States
| | - Abdullah Esmail
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX, United States
| | - Saifudeen Abdelrahim
- Challenge Early College HS, Houston Community College, Houston, TX, United States
| | - Karen Abboud
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX, United States
| | | | - Godsfavour Umoru
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX, United States
| | - Hashem A Rayyan
- Department of Medicine, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Ala Abudayyeh
- Section of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Maen Abdelrahim
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
28
|
Arayici ME, İnal A, Basbinar Y, Olgun N. Evaluation of the diagnostic and prognostic clinical values of circulating tumor DNA and cell-free DNA in pancreatic malignancies: a comprehensive meta-analysis. Front Oncol 2024; 14:1382369. [PMID: 38983931 PMCID: PMC11231086 DOI: 10.3389/fonc.2024.1382369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/10/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND The diagnostic and prognostic clinical value of circulating tumor DNA (ctDNA) and cell-free DNA (cfDNA) in pancreatic malignancies are unclear. Herein, we aimed to perform a meta-analysis to evaluate ctDNA and cfDNA as potential diagnostic and prognostic biomarkers. METHODS PRISMA reporting guidelines were followed closely for conducting the current meta-analysis. The PubMed/Medline, Scopus, and Web of Science (WoS) databases were scanned in detail to identify eligible papers for the study. A quality assessment was performed in accordance with the REMARK criteria. The risk ratios (RRs) of the diagnostic accuracy of ctDNA compared to that of carbohydrate antigen 19.9 (CA 19.9) in all disease stages and the hazard ratios (HRs) of the prognostic role of ctDNA in overall survival (OS) were calculated with 95% confidence intervals (CIs). RESULTS A total of 18 papers were evaluated to assess the diagnostic accuracy and prognostic value of biomarkers related to pancreatic malignancies. The pooled analysis indicated that CA19.9 provides greater diagnostic accuracy across all disease stages than ctDNA or cfDNA (RR = 0.64, 95% CI: 0.50-0.82, p < 0.001). Additionally, in a secondary analysis focusing on prognosis, patients who were ctDNA-positive were found to have significantly worse OS (HR = 2.00, 95% CI: 1.51-2.66, p < 0.001). CONCLUSION The findings of this meta-analysis demonstrated that CA19-9 still has greater diagnostic accuracy across all disease stages than KRAS mutations in ctDNA or cfDNA. Nonetheless, the presence of detectable levels of ctDNA was associated with worse patient outcomes regarding OS. There is a growing need for further research on this topic. SYSTEMATIC REVIEW REGISTRATION https://doi.org/10.37766/inplasy2023.12.0092, identifier INPLASY2023120092.
Collapse
Affiliation(s)
- Mehmet Emin Arayici
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Dokuz Eylül University, İzmir, Türkiye
- Department of Public Health, Faculty of Medicine, Dokuz Eylül University, İzmir, Türkiye
| | - Abdullah İnal
- Department of General Surgery, Faculty of Medicine, İzmir Democracy University, İzmir, Türkiye
| | - Yasemin Basbinar
- Department of Translational Oncology, Institute of Oncology, Dokuz Eylül University, İzmir, Türkiye
| | - Nur Olgun
- Department of Clinical Oncology, Institute of Oncology, Dokuz Eylül University, İzmir, Türkiye
| |
Collapse
|
29
|
Wang Y, Liu Y, Liu S, Cheng L, Liu X. Recent advances in N-glycan biomarker discovery among human diseases. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1156-1171. [PMID: 38910518 PMCID: PMC11464920 DOI: 10.3724/abbs.2024101] [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: 04/22/2024] [Accepted: 05/23/2024] [Indexed: 06/25/2024] Open
Abstract
N-glycans play important roles in a variety of biological processes. In recent years, analytical technologies with high resolution and sensitivity have advanced exponentially, enabling analysts to investigate N-glycomic changes in different states. Specific glycan and glycosylation signatures have been identified in multiple diseases, including cancer, autoimmune diseases, nervous system disorders, and metabolic and cardiovascular diseases. These glycans demonstrate comparable or superior indicating capability in disease diagnosis and prognosis over routine biomarkers. Moreover, synchronous glycan alterations concurrent with disease initiation and progression provide novel insights into pathogenetic mechanisms and potential treatment targets. This review elucidates the biological significance of N-glycans, compares the existing glycomic technologies, and delineates the clinical performance of N-glycans across a range of diseases.
Collapse
Affiliation(s)
- Yi Wang
- Department of Laboratory MedicineTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Yuanyuan Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key LaboratorySystems Biology ThemeDepartment of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074China
| | - Si Liu
- Department of Epidemiology and Health StatisticsSchool of Public HealthFujian Medical UniversityFuzhou350122China
| | - Liming Cheng
- Department of Laboratory MedicineTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Xin Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key LaboratorySystems Biology ThemeDepartment of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074China
| |
Collapse
|
30
|
Said SA, Perlmutter BC, Wehrle CJ, Chang J, Hossain MS, Naffouje S, Joyce D, Simon R, Walsh RM, Augustin T. Tumor Size Combined With CA-19 Level Improves Prediction of Survival of Patients With Pancreatic Adenocarcinoma Undergoing Perioperative Chemotherapy and Resection. Am Surg 2024; 90:1397-1405. [PMID: 38513242 DOI: 10.1177/00031348241241738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
BACKGROUND AND OBJECTIVE Five-year survival in pancreatic adenocarcinoma is less than 20%. While previous studies have postulated that a carbohydrate antigen 19-9 (CA19-9) threshold could predict outcome of resection, the role for CA19-9 in decision-making remains unclear. This study aims to assess whether CA19-9 levels combined with tumor size improve prediction of post-resection survival. METHOD A retrospective analysis was conducted on 109 patients with pancreatic adenocarcinoma who underwent perioperative chemotherapy followed by resection. The primary outcome of mortality was, divided into short (<1 year) or prolonged (>2 years). Univariate and multivariable analyses compared the tumor size-adjusted CA19-9 between the outcome groups. RESULTS Twenty-seven (24.78%) and eighty-two (75.23%) patients were in the short survival and prolonged-survival groups, respectively. The mean CA19-9 was significantly greater in the short vs prolonged group (P < .001). Analyzing CA19-9 level by tumor size, the association of high CA19-9 and short survival was significant for small (≤2 cm) and large tumor (>4 cm), but not for intermediate-size tumors (2-4 cm). Adjusting for preoperative variable did not change this association. CONCLUSION CA 19-9 in combination with tumor size better identifies patients with prolonged post-resection survival. This prediction is most accurate in patients with either small (≤2 cms) or large (>4 cms) tumors compared to intermediate-size tumors.
Collapse
Affiliation(s)
- Sayf A Said
- Department of General Surgery, Cleveland Clinic, Cleveland, OH, USA
| | | | - Chase J Wehrle
- Department of General Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Jenny Chang
- Department of General Surgery, Cleveland Clinic, Cleveland, OH, USA
| | | | - Samer Naffouje
- Department of General Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Daniel Joyce
- Department of General Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Robert Simon
- Department of General Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - R Matthew Walsh
- Department of General Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Toms Augustin
- Department of General Surgery, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
31
|
Thomas DS, Sebastian A, Thomas V, Thomas A, Chandy RG, Peedicayil A. Diagnostic Accuracy of CA 19-9 in Ovarian Malignancy. INDIAN JOURNAL OF GYNECOLOGIC ONCOLOGY 2024; 22:73. [DOI: 10.1007/s40944-024-00829-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/24/2024] [Accepted: 04/07/2024] [Indexed: 01/03/2025]
|
32
|
Xu ZJ, Li JA, Cao ZY, Xu HX, Ying Y, Xu ZH, Liu RJ, Guo Y, Zhang ZX, Wang WQ, Liu L. Construction of S100 family members prognosis prediction model and analysis of immune microenvironment landscape at single-cell level in pancreatic adenocarcinoma: a tumor marker prognostic study. Int J Surg 2024; 110:3591-3605. [PMID: 38498399 PMCID: PMC11175822 DOI: 10.1097/js9.0000000000001293] [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: 10/16/2023] [Accepted: 02/22/2024] [Indexed: 03/20/2024]
Abstract
Pancreatic adenocarcinoma characterized by a mere 10% 5-year survival rate, poses a formidable challenge due to its specific anatomical location, making tumor tissue acquisition difficult. This limitation underscores the critical need for novel biomarkers to stratify this patient population. Accordingly, this study aimed to construct a prognosis prediction model centered on S100 family members. Leveraging six S100 genes and their corresponding coefficients, an S100 score was calculated to predict survival outcomes. The present study provided comprehensive internal and external validation along with power evaluation results, substantiating the efficacy of the proposed model. Additionally, the study explored the S100-driven potential mechanisms underlying malignant progression. By comparing immune cell infiltration proportions in distinct patient groups with varying prognoses, the research identified differences driven by S100 expression. Furthermore, the analysis explored significant ligand-receptor pairs between malignant cells and immune cells influenced by S100 genes, uncovering crucial insights. Notably, the study identified a novel biomarker capable of predicting the sensitivity of neoadjuvant chemotherapy, offering promising avenues for further research and clinical application.
Collapse
Affiliation(s)
- Zi-jin Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
- Department of General Surgery, QingPu Branch of Zhongshan Hospital, Fudan University
| | - Jian-ang Li
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Ze-yuan Cao
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, People’s Republic of China
| | - Hua-xiang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Ying Ying
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Zhi-hang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Run-jie Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Yuquan Guo
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Zi-xin Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Wen-quan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| |
Collapse
|
33
|
Haab B, Qian L, Staal B, Jain M, Fahrmann J, Worthington C, Prosser D, Velokokhatnaya L, Lopez C, Tang R, Hurd MW, Natarajan G, Kumar S, Smith L, Hanash S, Batra SK, Maitra A, Lokshin A, Huang Y, Brand RE. A Rigorous Multi-Laboratory Study of Known PDAC Biomarkers Identifies Increased Sensitivity and Specificity Over CA19-9 Alone. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595399. [PMID: 38826212 PMCID: PMC11142185 DOI: 10.1101/2024.05.22.595399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
A blood test that enables surveillance for early-stage pancreatic ductal adenocarcinoma (PDAC) is an urgent need. Independent laboratories have reported PDAC biomarkers that could improve biomarker performance over CA19-9 alone, but the performance of the previously reported biomarkers in combination is not known. Therefore, we conducted a coordinated case/control study across multiple laboratories using common sets of blinded training and validation samples (132 and 295 plasma samples, respectively) from PDAC patients and non-PDAC control subjects representing conditions under which surveillance occurs. We analyzed the training set to identify candidate biomarker combination panels using biomarkers across laboratories, and we applied the fixed panels to the validation set. The panels identified in the training set, CA19-9 with CA199.STRA, LRG1, TIMP-1, TGM2, THSP2, ANG, and MUC16.STRA, achieved consistent performance in the validation set. The panel of CA19-9 with the glycan biomarker CA199.STRA improved sensitivity from 0.44 with 0.98 specificity for CA19-9 alone to 0.71 with 0.98 specificity (p < 0.001, 1000-fold bootstrap). Similarly, CA19-9 combined with the protein biomarker LRG1 and CA199.STRA improved specificity from 0.16 with 0.94 sensitivity for CA19-9 to 0.65 with 0.89 sensitivity (p < 0.001, 1000-fold bootstrap). We further validated significantly improved performance using biomarker panels that did not include CA19-9. This study establishes the effectiveness of a coordinated study of previously discovered biomarkers and identified panels of those biomarkers that significantly increased the sensitivity and specificity of early-stage PDAC detection in a rigorous validation trial.
Collapse
|
34
|
Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, Tian X, Guan X, Cen X, Zhao Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther 2024; 9:132. [PMID: 38763973 PMCID: PMC11102923 DOI: 10.1038/s41392-024-01823-2] [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: 06/05/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 05/21/2024] Open
Abstract
Tumor biomarkers, the substances which are produced by tumors or the body's responses to tumors during tumorigenesis and progression, have been demonstrated to possess critical and encouraging value in screening and early diagnosis, prognosis prediction, recurrence detection, and therapeutic efficacy monitoring of cancers. Over the past decades, continuous progress has been made in exploring and discovering novel, sensitive, specific, and accurate tumor biomarkers, which has significantly promoted personalized medicine and improved the outcomes of cancer patients, especially advances in molecular biology technologies developed for the detection of tumor biomarkers. Herein, we summarize the discovery and development of tumor biomarkers, including the history of tumor biomarkers, the conventional and innovative technologies used for biomarker discovery and detection, the classification of tumor biomarkers based on tissue origins, and the application of tumor biomarkers in clinical cancer management. In particular, we highlight the recent advancements in biomarker-based anticancer-targeted therapies which are emerging as breakthroughs and promising cancer therapeutic strategies. We also discuss limitations and challenges that need to be addressed and provide insights and perspectives to turn challenges into opportunities in this field. Collectively, the discovery and application of multiple tumor biomarkers emphasized in this review may provide guidance on improved precision medicine, broaden horizons in future research directions, and expedite the clinical classification of cancer patients according to their molecular biomarkers rather than organs of origin.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Tao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiahao Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyu Yang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yu Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- School of Medicine, Tibet University, Lhasa, 850000, China
| | - Xinyu Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinqi Guan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaobo Cen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
35
|
Shi X, Guo S, Duan Q, Zhang W, Gao S, Jing W, Jiang G, Kong X, Li P, Li Y, Teng C, Xu X, Chen S, Nian B, Li Z, Zhong C, Yang X, Zhu G, Du Y, Zhang D, Jin G. Detection and characterization of pancreatic and biliary tract cancers using cell-free DNA fragmentomics. J Exp Clin Cancer Res 2024; 43:145. [PMID: 38750539 PMCID: PMC11094938 DOI: 10.1186/s13046-024-03067-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Plasma cell-free DNA (cfDNA) fragmentomics has demonstrated significant differentiation power between cancer patients and healthy individuals, but little is known in pancreatic and biliary tract cancers. The aim of this study is to characterize the cfDNA fragmentomics in biliopancreatic cancers and develop an accurate method for cancer detection. METHODS One hundred forty-seven patients with biliopancreatic cancers and 71 non-cancer volunteers were enrolled, including 55 patients with cholangiocarcinoma, 30 with gallbladder cancer, and 62 with pancreatic cancer. Low-coverage whole-genome sequencing (median coverage: 2.9 ×) was performed on plasma cfDNA. Three cfDNA fragmentomic features, including fragment size, end motif and nucleosome footprint, were subjected to construct a stacked machine learning model for cancer detection. Integration of carbohydrate antigen 19-9 (CA19-9) was explored to improve model performance. RESULTS The stacked model presented robust performance for cancer detection (area under curve (AUC) of 0.978 in the training cohort, and AUC of 0.941 in the validation cohort), and remained consistent even when using extremely low-coverage sequencing depth of 0.5 × (AUC: 0.905). Besides, our method could also help differentiate biliopancreatic cancer subtypes. By integrating the stacked model and CA19-9 to generate the final detection model, a high accuracy in distinguishing biliopancreatic cancers from non-cancer samples with an AUC of 0.995 was achieved. CONCLUSIONS Our model demonstrated ultrasensitivity of plasma cfDNA fragementomics in detecting biliopancreatic cancers, fulfilling the unmet accuracy of widely-used serum biomarker CA19-9, and provided an affordable way for accurate noninvasive biliopancreatic cancer screening in clinical practice.
Collapse
Affiliation(s)
- Xiaohan Shi
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Shiwei Guo
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Qiaonan Duan
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China
| | - Wei Zhang
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China
| | - Suizhi Gao
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Wei Jing
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Guojuan Jiang
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China
| | - Xiangyu Kong
- Department of Gastroenterology, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Penghao Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yikai Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Chuanqi Teng
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xiaoya Xu
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China
| | - Sheng Chen
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China
| | - Baoning Nian
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China
| | - Zhikuan Li
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China
| | - Chaoliang Zhong
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xiaolu Yang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Guangyu Zhu
- Department of Interventional Radiology and Vascular Surgery, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu Province, 210009, China.
| | - Yiqi Du
- Department of Gastroenterology, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Dadong Zhang
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China.
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
36
|
Kikuchi Y, Shimada H, Yamasaki F, Yamashita T, Araki K, Horimoto K, Yajima S, Yashiro M, Yokoi K, Cho H, Ehira T, Nakahara K, Yasuda H, Isobe K, Hayashida T, Hatakeyama S, Akakura K, Aoki D, Nomura H, Tada Y, Yoshimatsu Y, Miyachi H, Takebayashi C, Hanamura I, Takahashi H. Clinical practice guidelines for molecular tumor marker, 2nd edition review part 2. Int J Clin Oncol 2024; 29:512-534. [PMID: 38493447 DOI: 10.1007/s10147-024-02497-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/21/2024] [Indexed: 03/19/2024]
Abstract
In recent years, rapid advancement in gene/protein analysis technology has resulted in target molecule identification that may be useful in cancer treatment. Therefore, "Clinical Practice Guidelines for Molecular Tumor Marker, Second Edition" was published in Japan in September 2021. These guidelines were established to align the clinical usefulness of external diagnostic products with the evaluation criteria of the Pharmaceuticals and Medical Devices Agency. The guidelines were scoped for each tumor, and a clinical questionnaire was developed based on a serious clinical problem. This guideline was based on a careful review of the evidence obtained through a literature search, and recommendations were identified following the recommended grades of the Medical Information Network Distribution Services (Minds). Therefore, this guideline can be a tool for cancer treatment in clinical practice. We have already reported the review portion of "Clinical Practice Guidelines for Molecular Tumor Marker, Second Edition" as Part 1. Here, we present the English version of each part of the Clinical Practice Guidelines for Molecular Tumor Marker, Second Edition.
Collapse
Affiliation(s)
| | - Hideaki Shimada
- Department of Clinical Oncology, Toho University, Tokyo, Japan.
- Department of Surgery, Toho University, Tokyo, Japan.
| | - Fumiyuki Yamasaki
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Taku Yamashita
- Department of Otorhinolaryngology-Head and Neck Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Koji Araki
- Department of Otorhinolaryngology-Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - Kohei Horimoto
- Department of Dermatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | - Masakazu Yashiro
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Keigo Yokoi
- Department of Lower Gastrointestinal Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Haruhiko Cho
- Department of Surgery, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | - Takuya Ehira
- Department of Gastroenterology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kazunari Nakahara
- Department of Gastroenterology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Hiroshi Yasuda
- Department of Gastroenterology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kazutoshi Isobe
- Division of Respiratory Medicine, Department of Internal Medicine (Omori), Toho University, Tokyo, Japan
| | - Tetsu Hayashida
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Shingo Hatakeyama
- Department of Urology, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | | | - Daisuke Aoki
- International University of Health and Welfare Graduate School, Tokyo, Japan
| | - Hiroyuki Nomura
- Department of Obstetrics and Gynecology, School of Medicine, Fujita Health University, Aichi, Japan
| | - Yuji Tada
- Department of Pulmonology, School of Medicine, International University of Health and Welfare, Chiba, Japan
| | - Yuki Yoshimatsu
- Department of Patient-Derived Cancer Model, Tochigi Cancer Center Research Institute, Tochigi, Japan
| | - Hayato Miyachi
- Faculty of Clinical Laboratory Sciences, Nitobe Bunka College, Tokyo, Japan
| | - Chiaki Takebayashi
- Division of Hematology and Oncology, Department of Internal Medicine (Omori), Toho University, Tokyo, Japan
| | - Ichiro Hanamura
- Division of Hematology, Department of Internal Medicine, Aichi Medical University, Aichi, Japan
| | | |
Collapse
|
37
|
Morikawa H, Oba T, Kitazawa A, Iji R, Kiyosawa N, Amitani M, Shimizu T, Kanai T, Uehara T, Ito KI. CA19-9 producing locally advanced papillary thyroid carcinoma: a case report. Surg Case Rep 2024; 10:83. [PMID: 38598167 PMCID: PMC11006640 DOI: 10.1186/s40792-024-01887-w] [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: 01/17/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND CA19-9 is a tumor marker for gastrointestinal and biliary-pancreatic adenocarcinomas; however, its association with thyroid cancer is unknown. Here, we report a case of CA19-9 producing locally advanced papillary thyroid carcinoma (PTC). CASE PRESENTATION A 66-year-old woman who was identified with a thyroid tumor after a close examination of an elevated serum CA19-9 level, which was detected at health screening, was referred to our hospital. Ultrasonography revealed a 34 × 31 mm hypoechoic lesion in the lower pole of the left thyroid lobe. Computed tomography revealed a solid thyroid tumor with tracheal invasion without any distant metastases. Bronchoscopy revealed tumor exposure into the tracheal lumen on the left side of the trachea. Fine-needle aspiration cytology led to a diagnosis of papillary thyroid carcinoma (PTC). The patient underwent a total thyroidectomy, tracheal sleeve resection with end-to-end anastomosis, and lymph node dissection in the left cervical and superior mediastinal regions (D3c) with a reversed T-shaped upper sternotomy down to the third intercostal level. Histopathological analysis confirmed the diagnosis of PTC with tracheal invasion and no lymph node metastases (pT4a Ex2 N0). Immunohistochemical staining showed the expression of CA19-9 in cancer cells. Postoperatively, the serum CA19-9 level of the patient decreased to within the normal range. CONCLUSIONS Some PTCs produce CA19-9, although less frequently. When elevated serum CA19-9 levels are observed, PTC should be included in the differential diagnosis for further investigation.
Collapse
Affiliation(s)
- Hiroki Morikawa
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Takaaki Oba
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan.
| | - Ayaka Kitazawa
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Ryoko Iji
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Nami Kiyosawa
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Masatsugu Amitani
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Tadafumi Shimizu
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Toshiharu Kanai
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Takeshi Uehara
- Division of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Ken-Ichi Ito
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| |
Collapse
|
38
|
Ansari N, Ozgur SS, Besada D, Bittar N, Melki G, Badipatla K, Christian D, Cavanagh Y. Carbohydrate Antigen (CA 19-9) Surge: Unraveling the Enigma of Elevated Levels in the Setting of Benign Etiologies. Cureus 2024; 16:e57469. [PMID: 38699139 PMCID: PMC11065394 DOI: 10.7759/cureus.57469] [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: 04/02/2024] [Indexed: 05/05/2024] Open
Abstract
Carbohydrate antigen 19-9 (CA 19-9) is widely recognized as a tumor marker primarily associated with pancreatic cancer. However, its elevation in benign pancreaticobiliary conditions complicates its diagnostic utility. We present the case of a 39-year-old male with no significant medical history who presented with symptoms of abdominal pain, nausea, vomiting, and diarrhea. The initial diagnosis suggested viral enteritis, but the subsequent worsening of symptoms led to further investigation. Elevated white blood cell counts, bilirubin levels, and liver function tests prompted magnetic resonance cholangiopancreatography (MRCP), which revealed dilated bile ducts and acute cholecystitis. Following endoscopic retrograde cholangiopancreatography (ERCP), significant hemobilia was observed, raising suspicions of cholangiocarcinoma. Despite extensive investigations, including CT angiography, MRCP, and repeat ERCPs, no malignancy was detected. Remarkably, the CA 19-9 level was elevated to 904 U/mL after the initial ERCP and uptrended to 7380 U/mL. These levels, however, normalized to 13 U/mL within two weeks of discharge. While CA 19-9 is a valuable marker in the diagnosis of pancreatic cancer, its elevation in benign pancreaticobiliary conditions necessitates cautious interpretation. In our case, choledocolithasis, cholangitis, and biliary manipulation appeared to have contributed to a transiently elevated CA 19-9. Clinicians must consider the entire clinical context when evaluating elevated CA 19-9 levels to avoid misdiagnosis and ensure appropriate patient management.
Collapse
Affiliation(s)
- Nida Ansari
- Internal Medicine, St. Joseph's Regional Medical Center, Paterson, USA
| | - Sacide S Ozgur
- Internal Medicine, St. Joseph's Regional Medical Center, Paterson, USA
| | - Damian Besada
- Internal Medicine, St. George's University School of Medicine, Great River, USA
| | - Noor Bittar
- Internal Medicine, St. George's University School of Medicine, Great River, USA
| | - Gabriel Melki
- Gastroenterology, St. Joseph's Regional Medical Center, Paterson, USA
| | - Kanthi Badipatla
- Gastroenterology, St. Joseph's Regional Medical Center, Paterson, USA
| | | | - Yana Cavanagh
- Gastroenterology, St. Joseph's Regional Medical Center, Paterson, USA
| |
Collapse
|
39
|
Zhang Y, Zhang H, Liu B, Ning K. Highly accurate diagnosis of pancreatic cancer by integrative modeling using gut microbiome and exposome data. iScience 2024; 27:109294. [PMID: 38450156 PMCID: PMC10915599 DOI: 10.1016/j.isci.2024.109294] [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: 07/06/2023] [Revised: 10/07/2023] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
The noninvasive detection of pancreatic ductal adenocarcinoma (PDAC) remains an immense challenge. In this study, we proposed a robust, accurate, and noninvasive classifier, namely Multi-Omics Co-training Graph Convolutional Networks (MOCO-GCN). It achieved high accuracy (0.9 ± 0.06), F1 score (0.9± 0.07), and AUROC (0.89± 0.08), surpassing contemporary approaches. The performance of model was validated on an external cohort of German PDAC patients. Additionally, we discovered that the exposome may impact PDAC development through its complex interplay with gut microbiome by mediation analysis. For example, Fusobacterium hwasookii nucleatum, known for its ability to induce inflammatory responses, may serve as a mediator for the impact of rheumatoid arthritis on PDAC. Overall, our study sheds light on how exposome and microbiome in concert could contribute to PDAC development, and enable PDAC diagnosis with high fidelity and interpretability.
Collapse
Affiliation(s)
- Yuli Zhang
- School of Mathematics, Shandong University, Jinan 250200, Shandong, China
| | - Haohong Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Bingqiang Liu
- School of Mathematics, Shandong University, Jinan 250200, Shandong, China
| | - Kang Ning
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| |
Collapse
|
40
|
Wang H, Wang Y, Zhang D, Li P. Circulating nucleosomes as potential biomarkers for cancer diagnosis and treatment monitoring. Int J Biol Macromol 2024; 262:130005. [PMID: 38331061 DOI: 10.1016/j.ijbiomac.2024.130005] [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: 11/29/2023] [Revised: 01/03/2024] [Accepted: 02/04/2024] [Indexed: 02/10/2024]
Abstract
Nucleosomes play a crucial role in regulating gene expression through their composition and post-translational modifications. When cells die, intracellular endonucleases are activated and cleave chromatin into oligo- and mono-nucleosomes, which are then released into the body fluids. Studies have shown that the levels of nucleosomes are increased in serum and plasma in various cancer types, suggesting that analysis of circulating nucleosomes can provide an initial assessment of carcinogenesis. However, it should be noted that elevated serum nucleosome levels may not accurately diagnose certain tumor types, as increased cell death may occur in different pathological conditions. Nevertheless, detection of circulating nucleosomes and their histone modifications, along with specific tumor markers, can help diagnose certain types of cancer. Furthermore, monitoring changes in circulating nucleosome levels during chemotherapy or radiotherapy in patients with malignancies can provide valuable insights into clinical outcomes and therapeutic efficacy. The utilization of circulating nucleosomes as biomarkers is an exciting and emerging area of research, with the potential for early detection of various diseases and monitoring of treatment response. Integrating nucleosome-based biomarkers with existing ones may improve the specificity and sensitivity of current assays, offering the possibility of personalized precision medical treatment for patients.
Collapse
Affiliation(s)
- Huawei Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 1 Ningde Road, Qingdao 266073, China.
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 1 Ningde Road, Qingdao 266073, China.
| | - Dejiu Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 1 Ningde Road, Qingdao 266073, China.
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 1 Ningde Road, Qingdao 266073, China.
| |
Collapse
|
41
|
Ciesielka J, Jakimów K, Tekiela N, Peisert L, Kwaśniewska A, Kata D, Chudek J. Significantly Elevated CA 19-9 after COVID-19 Vaccination and Literature Review of Non-Cancerous Cases with CA 19-9 > 1000 U/mL. J Clin Med 2024; 13:1263. [PMID: 38592088 PMCID: PMC10932348 DOI: 10.3390/jcm13051263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND CA 19-9 is a commonly assessed tumor marker, considered characteristic of pancreatic ductal adenocarcinoma (PDAC) and biliary tract cancers; however, the positive predictive value of CA 19.9 is too low, and the usage of CA 19.9 as a screening tool in the healthy population remains controversial. METHODS The presented case illustrates a reversed diagnosis of highly elevated serum CA 19-9 levels in a 54-year-old female complaining of pain in the epigastric region, shortly after COVID-19 vaccination. Laboratory tests showed a significantly elevated level of the CA 19-9 marker (>12,000 U/mL, reference value: <37 U/mL) with normal pancreatic enzyme activity. The patient underwent imaging examination, which showed no abnormalities, except for increased pancreatic dimension and areas of fluid signal in the pancreas in magnetic resonance imaging (MRI), which may correspond to autoimmune pancreatitis (AIP). The patient remains asymptomatic with a recommendation for a follow-up MRI in 12 months. RESULTS A literature review conducted revealed multi-causal CA 19-9 increases above 1000 U/mL, including non-cancerous diseases of the lung, pancreas, liver, ovary, kidney, and others. The median concentration of CA 19-9 regardless of the cause of disease was 2810 U/mL (IQR ± 6895). The median CA 19-9 values in men and women were 3500 (IQR ± 10,050) and 2455 (IQR ± 3927), respectively, and differ significantly between the compared groups (p < 0.05). There was no difference between CA 19-9 values and the categorized cause of the increase. CONCLUSIONS Conducting differential diagnosis, it should not be forgotten that most international guidelines recommend the use of CA 19-9 only in conjunction with pathology of pancreas in radiological imaging; however, even such a combination can point the diagnostic pathway in the wrong direction. A highly elevated CA 19-9 level, typically associated with PDAC, may be the result of benign disease including AIP related to COVID-19 vaccination.
Collapse
Affiliation(s)
- Jakub Ciesielka
- Student’s Research Group, Department of Internal Medicine and Oncological Chemotherapy, Medical University of Silesia in Katowice, 40-055 Katowice, Poland; (K.J.); (N.T.); (L.P.)
| | - Krzysztof Jakimów
- Student’s Research Group, Department of Internal Medicine and Oncological Chemotherapy, Medical University of Silesia in Katowice, 40-055 Katowice, Poland; (K.J.); (N.T.); (L.P.)
| | - Natalia Tekiela
- Student’s Research Group, Department of Internal Medicine and Oncological Chemotherapy, Medical University of Silesia in Katowice, 40-055 Katowice, Poland; (K.J.); (N.T.); (L.P.)
| | - Laura Peisert
- Student’s Research Group, Department of Internal Medicine and Oncological Chemotherapy, Medical University of Silesia in Katowice, 40-055 Katowice, Poland; (K.J.); (N.T.); (L.P.)
| | - Anna Kwaśniewska
- Department of Radiology, The Mielecki Hospital, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Dariusz Kata
- Department of Hematology and Bone Marrow Transplantation, Medical University of Silesia in Katowice, 40-055 Katowice, Poland;
| | - Jerzy Chudek
- Department of Internal Medicine and Oncological Chemotherapy, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| |
Collapse
|
42
|
Joseph AM, Al Aiyan A, Al-Ramadi B, Singh SK, Kishore U. Innate and adaptive immune-directed tumour microenvironment in pancreatic ductal adenocarcinoma. Front Immunol 2024; 15:1323198. [PMID: 38384463 PMCID: PMC10879611 DOI: 10.3389/fimmu.2024.1323198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/11/2024] [Indexed: 02/23/2024] Open
Abstract
One of the most deadly and aggressive cancers in the world, pancreatic ductal adenocarcinoma (PDAC), typically manifests at an advanced stage. PDAC is becoming more common, and by the year 2030, it is expected to overtake lung cancer as the second greatest cause of cancer-related death. The poor prognosis can be attributed to a number of factors, including difficulties in early identification, a poor probability of curative radical resection, limited response to chemotherapy and radiotherapy, and its immunotherapy resistance. Furthermore, an extensive desmoplastic stroma that surrounds PDAC forms a mechanical barrier that prevents vascularization and promotes poor immune cell penetration. Phenotypic heterogeneity, drug resistance, and immunosuppressive tumor microenvironment are the main causes of PDAC aggressiveness. There is a complex and dynamic interaction between tumor cells in PDAC with stromal cells within the tumour immune microenvironment. The immune suppressive microenvironment that promotes PDAC aggressiveness is contributed by a range of cellular and humoral factors, which itself are modulated by the cancer. In this review, we describe the role of innate and adaptive immune cells, complex tumor microenvironment in PDAC, humoral factors, innate immune-mediated therapeutic advances, and recent clinical trials in PDAC.
Collapse
Affiliation(s)
- Ann Mary Joseph
- Department of Veterinary Medicine (CAVM), United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ahmad Al Aiyan
- Department of Veterinary Medicine (CAVM), United Arab Emirates University, Al Ain, United Arab Emirates
| | - Basel Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Shiv K. Singh
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center, Goettingen, Germany
| | - Uday Kishore
- Department of Veterinary Medicine (CAVM), United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
43
|
Yousef A, Yousef M, Zeineddine MA, More A, Fanaeian M, Chowdhury S, Knafl M, Edelkamp P, Ito I, Gu Y, Pattalachinti V, Naini ZA, Zeineddine FA, Peterson J, Alfaro K, Foo WC, Jin J, Bhutiani N, Higbie V, Scally CP, Kee B, Kopetz S, Goldstein D, Strach M, Williamson A, Aziz O, Barriuso J, Uppal A, White MG, Helmink B, Fournier KF, Raghav KP, Taggart MW, Overman MJ, Shen JP. Serum Tumor Markers and Outcomes in Patients With Appendiceal Adenocarcinoma. JAMA Netw Open 2024; 7:e240260. [PMID: 38416491 PMCID: PMC10902735 DOI: 10.1001/jamanetworkopen.2024.0260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/02/2024] [Indexed: 02/29/2024] Open
Abstract
Importance Serum tumor markers carcinoembryonic antigen (CEA), carbohydrate antigen 19-9 (CA19-9), and cancer antigen 125 (CA125) have been useful in the management of gastrointestinal and gynecological cancers; however, there is limited information regarding their utility in patients with appendiceal adenocarcinoma. Objective To assess the association of serum tumor markers (CEA, CA19-9, and CA125) with clinical outcomes and pathologic and molecular features in patients with appendiceal adenocarcinoma. Design, Setting, and Participants This is a retrospective cohort study at a single tertiary care comprehensive cancer center. The median (IQR) follow-up time was 52 (21-101) months. Software was used to query the MD Anderson internal patient database to identify patients with a diagnosis of appendiceal adenocarcinoma and at least 1 tumor marker measured at MD Anderson between March 2016 and May 2023. Data were analyzed from January to December 2023. Main Outcomes and Measures Association of serum tumor markers with survival in patients with appendiceal adenocarcinoma. Cox proportional hazards regression analyses were also performed to assess associations between clinical factors (serum tumor marker levels, demographics, and patient and disease characteristics) and patient outcomes (overall survival). Results A total of 1338 patients with appendiceal adenocarcinoma were included, with a median (range) age at diagnosis of 56.5 (22.3-89.6) years. The majority of the patients had metastatic disease (1080 patients [80.7%]). CEA was elevated in 742 of the patients tested (56%), while CA19-9 and CA125 were elevated in 381 patients (34%) and 312 patients (27%), respectively. Individually, elevation of CEA, CA19-9, or CA125 were associated with worse 5-year survival; elevated vs normal was 81% vs 95% for CEA (hazard ratio [HR], 4.0; 95% CI, 2.9-5.6), 84% vs 92% for CA19-9 (HR, 2.2; 95% CI, 1.4-3.4), and 69% vs 93% for CA125 (HR, 4.6; 95% CI, 2.7-7.8) (P < .001 for all). Quantitative evaluation of tumor markers was associated with outcomes. Patients with highly elevated (top 10th percentile) CEA, CA19-9, or CA125 had markedly worse survival, with 5-year survival rates of 59% for CEA (HR, 9.8; 95% CI, 5.3-18.0), 64% for CA19-9 (HR, 6.0; 95% CI, 3.0-11.7), and 57% for CA125 (HR, 7.6; 95% CI, 3.5-16.5) (P < .001 for all). Although metastatic tumors had higher levels of all tumor markers, when restricting survival analysis to 1080 patients with metastatic disease, elevated CEA, CA19-9, or CA125 were all still associated worse survival (HR for CEA, 3.4; 95% CI, 2.5-4.8; P < .001; HR for CA19-9, 1.8; 95% CI, 1.2-2.7; P = .002; and HR for CA125, 3.9; 95% CI, 2.4-6.4; P < .001). Interestingly, tumor grade was not associated with CEA or CA19-9 level, while CA-125 was slightly higher in high-grade tumors relative to low-grade tumors (mean value, 18.3 vs 15.0; difference, 3.3; 95% CI, 0.9-3.7; P < .001). Multivariable analysis identified an incremental increase in the risk of death with an increase in the number of elevated tumor markers, with an 11-fold increased risk of death in patients with all 3 tumor markers elevated relative to those with none elevated. Somatic mutations in KRAS and GNAS were associated with significantly higher levels of CEA and CA19-9. Conclusions and Relevance In this retrospective study of serum tumor markers in patients with appendiceal adenocarcinoma, CEA, CA19-9, and CA125 were associated with overall survival in appendiceal adenocarcinoma. Given their value, all 3 biomarkers should be included in the initial workup of patients with a diagnosis of appendiceal adenocarcinoma.
Collapse
Affiliation(s)
- Abdelrahman Yousef
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Mohammad A. Zeineddine
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Aditya More
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Mohammad Fanaeian
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Saikat Chowdhury
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Mark Knafl
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston
| | - Paul Edelkamp
- Department of Data Engineering and Analytics, University of Texas MD Anderson Cancer Center, Houston
| | - Ichiaki Ito
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Yue Gu
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Vinay Pattalachinti
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Zahra Alavi Naini
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Fadl A. Zeineddine
- Department of Internal Medicine, Houston Methodist Hospital, Houston, Texas
| | - Jennifer Peterson
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Kristin Alfaro
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Wai Chin Foo
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston
| | - Jeff Jin
- Department of Enterprise Development and Integration, University of Texas MD Anderson Cancer Center, Houston
| | - Neal Bhutiani
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston
| | - Victoria Higbie
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Christopher P. Scally
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Bryan Kee
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | | | - Madeleine Strach
- Colorectal and Peritoneal Oncology Centre, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
- Faculty of Medicine and Health, The University of Sydney, Darlington, Victoria, Australia
| | - Andrew Williamson
- Department of Medical Oncology, The Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Omer Aziz
- Colorectal and Peritoneal Oncology Centre, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Jorge Barriuso
- Colorectal and Peritoneal Oncology Centre, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Abhineet Uppal
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston
| | - Michael G. White
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston
| | - Beth Helmink
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Keith F. Fournier
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Kanwal P. Raghav
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Melissa W. Taggart
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston
| | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
44
|
Ahmadipour M, Bhattacharya A, Sarafbidabad M, Syuhada Sazali E, Krishna Ghoshal S, Satgunam M, Singh R, Rezaei Ardani M, Missaoui N, Kahri H, Pal U, Ling Pang A. CA19-9 and CEA biosensors in pancreatic cancer. Clin Chim Acta 2024; 554:117788. [PMID: 38246211 DOI: 10.1016/j.cca.2024.117788] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/15/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
Cancer is a complex pathophysiological condition causing millions of deaths each year. Early diagnosis is essential especially for pancreatic cancer. Existing diagnostic tools rely on circulating biomarkers such as Carbohydrate Antigen 19-9 (CA19-9) and Carcinoembryonic Antigen (CEA). Unfortunately, these markers are nonspecific and may be increased in a variety of disorders. Accordingly, diagnosis of pancreatic cancer generally involves more invasive approaches such as biopsy as well as imaging studies. Recent advances in biosensor technology have allowed the development of precise diagnostic tools having enhanced analytical sensitivity and specificity. Herein we examine these advances in the detection of cancer in general and in pancreatic cancer specifically. Furthermore, we highlight novel technologies in the measurement of CA19-9 and CEA and explore their future application in the early detection of pancreatic cancer.
Collapse
Affiliation(s)
- Mohsen Ahmadipour
- Institute of Power Engineering, Universiti Tenaga Nasional, 43650 Serdang, Selangor, Malaysia.
| | - Anish Bhattacharya
- Advanced Optical Materials Research Group, Department of Physics, Faculty of Science, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia; Ibnu Sina Institute of Laser Centre, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia
| | - Mohsen Sarafbidabad
- Biomedical Engineering Department, Faculty of Engineering, University of Isfahan, Isfahan, Iran
| | - Ezza Syuhada Sazali
- Advanced Optical Materials Research Group, Department of Physics, Faculty of Science, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia; Ibnu Sina Institute of Laser Centre, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia
| | - Sib Krishna Ghoshal
- Advanced Optical Materials Research Group, Department of Physics, Faculty of Science, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia; Ibnu Sina Institute of Laser Centre, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia
| | - Meenaloshini Satgunam
- Institute of Power Engineering, Universiti Tenaga Nasional, 43650 Serdang, Selangor, Malaysia; Department of Mechanical Engineering, Universiti Tenaga Nasional, 43650 Serdang, Selangor, Malaysia
| | - Ramesh Singh
- Institute of Power Engineering, Universiti Tenaga Nasional, 43650 Serdang, Selangor, Malaysia; Center of Advanced Manufacturing and Materials Processing (AMMP), Faculty of Engineering, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Mohammad Rezaei Ardani
- School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, Engineering Campus, 14300 Nibong Tebal, Pulau Pinang, Malaysia
| | - Nadhem Missaoui
- Laboratory of Interfaces and Advanced Materials, Faculty of Sciences, University of Monastir, Monastir, Tunisia
| | - Hamza Kahri
- Laboratory of Interfaces and Advanced Materials, Faculty of Sciences, University of Monastir, Monastir, Tunisia
| | - Ujjwal Pal
- Department of Analytical and Structural Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Ai Ling Pang
- Department of Chemical Science, Faculty of Science, Universiti Tunku Abdul Rahman, 31900 Kampar, Perak, Malaysia
| |
Collapse
|
45
|
Kuttanchettiyar KG, V VK, Chisthi MM. Cyfra 21.1: A Useful Tumour Marker in Pancreatic Ductal Adenocarcinoma: Cross-Sectional Study. Cancer Control 2024; 31:10732748241228037. [PMID: 38246662 PMCID: PMC10802135 DOI: 10.1177/10732748241228037] [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: 09/04/2023] [Revised: 10/30/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Pancreatic cancer is a challenging disease, often requiring invasive procedures for diagnosis. Reliable tumour markers are essential for ensuring early detection and better patient outcomes. Although Carbohydrate Antigen 19-9 is the most commonly used marker, it is marred by low predictive accuracy and high false positivity. Carcino Embryonic Antigen also has limited practical use. A novel antigen, Cytokeratin fragment 21-1, is gaining significance for its diagnostic value in various tumours. MATERIALS AND METHODS This prospective study aimed to evaluate the potential of Cytokeratin fragment 21-1 in comparison with Carbohydrate Antigen 19-9 and Carcino Embryonic Antigen in diagnosing pancreatic cancer. From January 2016 to December 2019, 45 patients with confirmed pancreatic ductal adenocarcinoma were included in this cross-sectional study. RESULTS Carbohydrate Antigen 19-9 was raised in 22 patients, Carcino Embryonic Antigen was elevated in 17, and Cytokeratin fragment 21-1 was elevated in 30 cases. Carbohydrate Antigen 19-9 was found to be elevated in the presence of jaundice. Both Carbohydrate Antigen 19-9 and Cytokeratin fragment 21-1 had good correlation with stage of cancer, while Carcino Embryonic Antigen had very minimal correlation. CONCLUSION In this study, Cytokeratin fragment 21-1 was elevated in a higher number of cases than Carbohydrate Antigen 19-9 and Carcino Embryonic Antigen. Both Cytokeratin fragment 21-1 and Carbohydrate Antigen 19-9 correlated well with cancer stage. Also Cytokeratin fragment 21-1 was not affected by jaundice, unlike Carbohydrate Antigen 19-9. Therefore, Cytokeratin fragment 21-1 has the potential to be an effective individual tumour marker in pancreatic cancer.
Collapse
Affiliation(s)
| | | | - Meer M. Chisthi
- Department of General Surgery, Government Medical College, Trivandrum, India
| |
Collapse
|
46
|
Johnston AJ, Sivakumar S, Zhou Y, Funston G, Bradley SH. Improving early diagnosis of pancreatic cancer in symptomatic patients. Br J Gen Pract 2023; 73:534-535. [PMID: 38035808 PMCID: PMC10688932 DOI: 10.3399/bjgp23x735585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023] Open
Affiliation(s)
| | - Shivan Sivakumar
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham
| | - Yin Zhou
- National Institute for Health and Care Research (NIHR) Academic Clinical Lecturer, Wolfson Institute of Population Health, Queen Mary University of London, London
| | - Garth Funston
- Wolfson Institute of Population Health, Queen Mary University of London, London
| | - Stephen H Bradley
- NIHR Academic Clinical Lecturer, Leeds Institute of Health Sciences, University of Leeds, Leeds
| |
Collapse
|
47
|
Tarasiuk A, Mirocha G, Fichna J. Current status of Complementary and Alternative Medicine Interventions in the Management of Pancreatic Cancer - An Overview. Curr Treat Options Oncol 2023; 24:1852-1869. [PMID: 38079061 PMCID: PMC10781793 DOI: 10.1007/s11864-023-01146-4] [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] [Accepted: 10/24/2023] [Indexed: 01/11/2024]
Abstract
OPINION STATEMENT Pancreatic cancer (PC) remains the deadliest cancer worldwide. Most patients are diagnosed at the advanced or metastatic stage, leading to a poor prognosis. Awareness of the limitations of current therapy and accompanying pain, depression, malnutrition, and side effects of chemoradiotherapy may lead patients and physicians towards complementary and alternative medicine (CAM). CAM refers to a diverse set of medical and healthcare practices, products, and systems that are not part of conventional Western medicine. Despite the low-quality evidence supporting the efficacy of these methods, they remain appealing due to patients' beliefs, fear of death, and the slow development of conventional therapy. Hence, the possibility of using natural products for pancreatic cancer is increasing. CAM options such as: medical cannabis, plants, fungi, herbal formulas, and injections, which originate primarily from traditional Chinese or Japanese medicine i.e. Curcuma longa, Panax ginseng, Poria cocos, Hochuekkito, Juzentaihoto, and Rikkunshito, Shi-quan-da-bu-tang/TJ-48, Huang-qin-tang, Shuangbai San, Wen Jing Zhi Tong Fang, Xiang-Sha-Liu-jun-zi-tang, Aidi injection, Brucea javanica oil emulsion/Yadanziyouru injection, Compound Kushen injection, Huachansu injection, Kangai injection and Kanglaite injections are becoming promising candidates for the management of pancreatic cancer. The abovementioned substances/medications are the most popular or potentially effective in PC treatment and consequently CAM-based adjuvant therapy through improving patients' quality of life, might be a useful addition in the treatment of pancreatic cancer patients.
Collapse
Affiliation(s)
- Aleksandra Tarasiuk
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland.
| | - Grzegorz Mirocha
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| |
Collapse
|
48
|
Li J, Lin J, Ji Y, Wang X, Fu D, Wang W, Shen B. A novel pyroptosis-associated lncRNA LINC01133 promotes pancreatic adenocarcinoma development via miR-30b-5p/SIRT1 axis. Cell Oncol (Dordr) 2023; 46:1381-1398. [PMID: 37138146 PMCID: PMC10618383 DOI: 10.1007/s13402-023-00818-5] [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] [Accepted: 04/16/2023] [Indexed: 05/05/2023] Open
Abstract
PURPOSE Pancreatic adenocarcinoma (PAAD) remains a highly aggressive gastrointestinal malignancy with a dismal prognosis. Pyroptosis has a key role in tumor development. Long noncoding RNAs (lncRNAs) are involved in tumorigenesis and pyroptosis regulation. However, the prognostic potential and function of pyroptosis-related lncRNAs (PRLs) in PAAD remain unclear. We aimed to identify PRLs with promising predictive value for PAAD prognosis and investigate the mechanism by which PRLs affect pyroptosis and PAAD development. METHODS Key genes that regulate pyroptosis were determined from previous studies, and PRLs were identified from lncRNAs shown to be co-expressed in The Cancer Genome Atlas. Cox analysis and the least absolute shrinkage and selection operator (LASSO) regression model was used to establish a prognostic PRL signature. The clinical significance and functional mechanisms of LINC01133 were explored in vitro and in vivo. RESULTS A seven-lncRNA signature was established and the high-risk subgroup exhibited a shorter survival time. With lower immune infiltration abundance, poor immune function, and higher tumor mutational burden (TMB), the high-risk subgroup reflected a more immunosuppressive status with a greater scope for benefiting from immunotherapy. After LINC01133 knockdown, PAAD cells showed lower viability and higher pyroptosis-related gene expression. LINC01133 functioned as a competing endogenous RNA to sequester miR-30b-5p from sponging SIRT1 mRNA to inhibit PAAD pyroptosis. CONCLUSION With significant prognostic value, our PRL signature are involved in the biological processes of PAAD cells and associated with the immune environment. LINC01133 suppresses pyroptosis to promote PAAD development and could serve as a potential target for PAAD treatment.
Collapse
Affiliation(s)
- Jingwei Li
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiewei Lin
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuchen Ji
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xuelong Wang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China.
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Da Fu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China.
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Weishen Wang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China.
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China.
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
49
|
Nicoletti A, Vitale F, Quero G, Paratore M, Fiorillo C, Negri M, Carlino A, Inzani F, Gasbarrini A, Alfieri S, Zileri Dal Verme L. Immunohistochemical Evaluation of the Expression of Specific Membrane Antigens in Patients with Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2023; 15:4586. [PMID: 37760554 PMCID: PMC10526869 DOI: 10.3390/cancers15184586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/26/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
(1) Background: Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies. The lack of validated disease biomarkers makes timely diagnosis challenging in most cases. Cell membrane and surface proteins play a crucial role in several routes of oncogenesis. The aim of this study was to evaluate the expression of six membrane antigens on PDAC (CA 19-9, mucin 1 and 4 (MUC1, MUC4), mesothelin (MSLN), Annexin A10 (ANXA10), Glypican-1 (GPC-1)) and their correlation with oncologic outcomes. (2) Methods: Immunohistochemical staining for CA 19.9, MUC1, MUC4, MSLN, ANXA10, and GPC-1 of surgical samples of 50 consecutive patients with PDAC was performed. Antigen expression for tumor, ductal, and acinar tissues was classified according to the histo-score (H-score) by two pathologists. (3) Results: Recurrence rate was 47% and 18 patients (36%) deceased (median follow-up 21.5 months). Immunostaining for CA 19-9 and MUC1 showed a significantly higher expression in the neoplastic tissue compared to non-tumor ductal and acinar tissues (p < 0.001). MUC4, MSLN, ANXA10, and GPC-1 were selectively expressed in the neoplastic tissue (p < 0.001). A CA 19-9 H-score value >270 was independently associated with a worse overall survival (p = 0.05) and disease-free survival (p = 0.05). (4) Conclusions: CA 19-9 and MUC1 are highly expressed in PDAC cells. The histological expression of CA 19-9 may predict prognosis. MUC4, MSLN, ANXA10, and GPC-1 are selectively expressed by neoplastic tissue and may represent a potential histological biomarker of disease.
Collapse
Affiliation(s)
- Alberto Nicoletti
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (F.V.); (M.P.); (M.N.); (L.Z.D.V.)
| | - Federica Vitale
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (F.V.); (M.P.); (M.N.); (L.Z.D.V.)
| | - Giuseppe Quero
- Chirurgia Digestiva, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (G.Q.); (C.F.); (S.A.)
| | - Mattia Paratore
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (F.V.); (M.P.); (M.N.); (L.Z.D.V.)
| | - Claudio Fiorillo
- Chirurgia Digestiva, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (G.Q.); (C.F.); (S.A.)
| | - Marcantonio Negri
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (F.V.); (M.P.); (M.N.); (L.Z.D.V.)
| | - Angela Carlino
- Anatomia Patologica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.C.); (F.I.)
| | - Frediano Inzani
- Anatomia Patologica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.C.); (F.I.)
| | - Antonio Gasbarrini
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (F.V.); (M.P.); (M.N.); (L.Z.D.V.)
| | - Sergio Alfieri
- Chirurgia Digestiva, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (G.Q.); (C.F.); (S.A.)
| | - Lorenzo Zileri Dal Verme
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (F.V.); (M.P.); (M.N.); (L.Z.D.V.)
| |
Collapse
|
50
|
Yousef A, Yousef M, Zeineddine M, More A, Chowdhury S, Knafl M, Edelkamp P, Ito I, Gu Y, Pattalachinti V, Naini ZA, Zeineddine F, Peterson J, Alfaro K, Foo WC, Jin J, Bhutiani N, Higbie V, Scally C, Kee B, Kopetz S, Goldstein D, Uppal A, White MG, Helmink B, Fournier K, Raghav K, Taggart M, Overman MJ, Shen JP. The Clinical Significance of CEA, CA19-9, and CA125 in Management of Appendiceal Adenocarcinoma. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.10.23295319. [PMID: 37745596 PMCID: PMC10516068 DOI: 10.1101/2023.09.10.23295319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Importance Serum tumor markers CEA, CA19-9, & CA125 have been useful in the management of gastrointestinal and gynecological cancers, however there is limited information regarding their utility in patients with appendiceal adenocarcinoma. Objective Assessing the association of serum tumor markers (CEA, CA19-9, and CA125) with clinical outcomes, pathologic, and molecular features in patients with appendiceal adenocarcinoma. Design This is a retrospective study with results reported in 2023. The median follow-up time was 43 months. Setting Single tertiary care comprehensive cancer center. Participants Under an approved Institutional Review Board protocol, the Palantir Foundry software system was used to query the MD Anderson internal patient database to identify patients with a diagnosis of appendiceal adenocarcinoma and at least one tumor marker measured at MD Anderson between 2016 and 2023. Results A total of 1,338 patients with appendiceal adenocarcinoma were included, with a median age of 56.5 years. The majority of the patients had metastatic disease (80.7%). CEA was elevated in more than half of the patients tested (56%), while CA19-9 and CA125 were elevated in 34% and 27%, respectively. Individually, elevation of CEA, CA19-9, or CA125 were associated with worse 5-year survival; 82% vs 95%, 84% vs 92%, and 69% vs 93% elevated vs normal for CEA, CA19-9, and CA125 respectively (all p<0.0001). Quantitative evaluation of tumor markers increased prognostic ability. Patients with highly elevated (top 10th percentile) CEA, CA19-9 or CA125 had markedly worse survival with 5-year survival rates of 59%, 64%, and 57%, respectively (HR vs. normal : 9.8, 6.0, 7.6, all p<0.0001). Although metastatic tumors had higher levels of all tumor markers, when restricting survival analysis to 1080 patients with metastatic disease elevated CEA, CA19-9 or CA125 were all still associated worse survival (HR vs. normal : 3.4, 1.8, 3.9, p<0.0001 for CEA and CA125, p=0.0019 for CA19-9). Interestingly tumor grade was not associated with CEA or CA19-9 level, while CA-125 was slightly higher in high relative to low-grade tumors (18.3 vs. 15.0, p=0.0009). Multivariable analysis identified an incremental increase in the risk of death with an increase in the number of elevated tumor markers, with a 11-fold increased risk of death in patients with all three tumor markers elevated relative to those with none elevated. Mutation in KRAS and GNAS were associated with significantly higher levels of CEA and CA19-9. Conclusions These findings demonstrate the utility of measuring CEA, CA19-9, and CA125 in the management of appendiceal adenocarcinoma. Given their prognostic value, all three biomarkers should be included in the initial workup of patients diagnosed with appendiceal adenocarcinoma.
Collapse
Affiliation(s)
- Abdelrahman Yousef
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mohammad Zeineddine
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aditya More
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Saikat Chowdhury
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark Knafl
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Edelkamp
- Department of Data Engineering & Analytics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ichiaki Ito
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yue Gu
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vinay Pattalachinti
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zahra Alavi Naini
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fadl Zeineddine
- Department of Internal Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Jennifer Peterson
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristin Alfaro
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wai Chin Foo
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeff Jin
- Department of Enterprise Dev & Integration, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neal Bhutiani
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Victoria Higbie
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher Scally
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bryan Kee
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Abhineet Uppal
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael G. White
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Beth Helmink
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keith Fournier
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kanwal Raghav
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melissa Taggart
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|