1
|
Balaraman AK, Moglad E, Afzal M, Babu MA, Goyal K, Roopashree R, Kaur I, Kumar S, Kumar MR, Chauhan AS, Hemalatha S, Gupta G, Ali H. Liquid biopsies and exosomal ncRNA: Transforming pancreatic cancer diagnostics and therapeutics. Clin Chim Acta 2025; 567:120105. [PMID: 39706249 DOI: 10.1016/j.cca.2024.120105] [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: 11/16/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Pancreatic cancer is a highly fatal malignancy due to poor early detection rate and resistance to conventional therapies. This review examines the potential for liquid biopsy as a transformative technology to identify diagnostic and therapeutic targets in pancreatic cancer. Specifically, we explore emerging biomarkers such as exosomal non-coding RNAs (ncRNAs), circulating tumor DNA (ctDNA), and circulating tumor cells (CTCs). Tumor-derived exosomes contain nucleic acid and protein that reflect the unique molecular landscape of the malignancy and can serve as an alternative diagnostic approach vs traditional biomarkers like CA19-9. Herein we highlight exosomal miRNAs, lncRNAs, and other ncRNAs alongside ctDNA and CTC-based strategies, evaluating their combined ability to improve early detection, disease monitoring and treatment response. Furthermore, the therapeutic implications of ncRNAs such as lncRNA UCA1 and miR-3960 in chemoresistance and progression are also discussed via suppression of EZH2 and PTEN/AKT pathways. Emerging therapeutic strategies that target the immune response, epithelial-mesenchymal transition (EMT) and drug resistance are explored. This review demonstrates a paradigm shift in pancreatic cancer management toward personalized, less invasive and more effective approaches.
Collapse
Affiliation(s)
- Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - MRavi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research and Innovation, Uttaranchal University, India
| | - S Hemalatha
- Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Porur, Chennai, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| |
Collapse
|
2
|
Choi Y, Park JH, Jo A, Lim CW, Park JM, Hwang JW, Lee KS, Kim YS, Lee H, Moon J. Blood-derived APLP1 + extracellular vesicles are potential biomarkers for the early diagnosis of brain diseases. SCIENCE ADVANCES 2025; 11:eado6894. [PMID: 39742488 DOI: 10.1126/sciadv.ado6894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025]
Abstract
The early detection of neurodegenerative diseases necessitates the identification of specific brain-derived biomolecules in peripheral blood. In this context, our investigation delineates the role of amyloid precursor-like protein 1 (APLP1)-a protein predominantly localized in oligodendrocytes and neurons-as a previously unidentified biomarker in extracellular vesicles (EVs). Through rigorous analysis, APLP1+ EVs from human sera were unequivocally determined to be of cerebral origin. This assertion was corroborated by distinctive small RNA expression patterns of APLP1+ EVs. The miRNAs' putative targets within these EVs manifested pronounced expression in the brain, fortifying their neurospecific provenance. We subjected our findings to stringent validation using Thy-1 GFP M line mice, transgenic models wherein GFP expression is confined to hippocampal neurons. An amalgamation of these results with an exhaustive data analysis accentuates the potential of APLP1+ EVs as cerebrally originated biomarkers. Synthesizing our findings, APLP1+ EVs are postulated not merely as diagnostic markers but as seminal entities shaping the future trajectory of neurodegenerative disease diagnostics.
Collapse
Affiliation(s)
- Yuri Choi
- Department of Biotechnology, College of Life Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Jae Hyun Park
- Department of Biotechnology, College of Life Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Ala Jo
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Chul-Woo Lim
- Department of Biotechnology, College of Life Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Ji-Min Park
- Department of Biotechnology, College of Life Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Jin Woo Hwang
- Department of Biotechnology, College of Life Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Kang Soo Lee
- Department of Psychiatry, CHA Bundang Medical Center, CHA University College of Medicine, Gyeonggi-do 13496, Republic of Korea
| | - Young-Sang Kim
- Department of Family Medicine, CHA Bundang Medical Center, CHA University College of Medicine, Gyeonggi-do 13496, Republic of Korea
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jisook Moon
- Department of Biotechnology, College of Life Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| |
Collapse
|
3
|
Thongyoo P, Chindaprasirt J, Aphivatanasiri C, Intarawichian P, Kunprom W, Kongpetch S, Techasen A, Loilome W, Namwat N, Titapun A, Jusakul A. KRAS Mutations in Cholangiocarcinoma: Prevalence, Prognostic Value, and KRAS G12/G13 Detection in Cell-Free DNA. Cancer Genomics Proteomics 2025; 22:112-126. [PMID: 39730186 PMCID: PMC11696325 DOI: 10.21873/cgp.20492] [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: 08/24/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND/AIM Cholangiocarcinoma (CCA) is an aggressive hepatobiliary malignancy characterized by genomic heterogeneity. KRAS mutations play a significant role in influencing patient prognosis and guiding therapeutic decision-making. This study aimed to determine the prevalence and prognostic significance of KRAS mutations in CCA, asses the detection of KRAS G12/G13 mutations in plasma cell-free DNA (cfDNA), and evaluate the prognostic value of KRAS G12/G13 mutant allele frequency (MAF) in cfDNA in relation to clinicopathological data and patient survival. MATERIALS AND METHODS A retrospective analysis of 937 CCA patients was performed using data from cBioPortal to examine KRAS mutation profiles and their association with survival. Plasma from 101 CCA patients was analyzed for KRAS G12/G13 mutations in the cfDNA using droplet digital PCR, and the results were compared with tissue-based sequencing from 78 matched samples. RESULTS KRAS driver mutations were found in 15.6% of patients, with common variants being G12D (37.0%), G12V (24.0%) and Q61H (8.2%). Patients harboring KRAS mutations exhibited decreased overall and recurrence-free survival. KRAS G12/G13 mutations were detected in 14.9% of cfDNA samples, showing moderate concordance with tissue sequencing, and achieving 80% sensitivity and 93% specificity. Elevated KRAS G12/G13 MAF in cfDNA, combined with high CA19-9 levels, correlated with poorer survival outcomes. CONCLUSION The presence of KRAS mutations was associated with poor survival in CCA, underscoring the importance of KRAS mutations as prognostic markers. The detection of KRAS mutations in cfDNA demonstrated potential as a promising non-invasive alternative for mutation detection and, when combined with CA19-9 levels, may improve prognostic efficacy in CCA.
Collapse
Affiliation(s)
- Pitchasak Thongyoo
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Jarin Chindaprasirt
- Medical Oncology Program, Department of Internal Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | | | | - Waritta Kunprom
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sarinya Kongpetch
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Anchalee Techasen
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Watcharin Loilome
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Nisana Namwat
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Attapol Titapun
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Apinya Jusakul
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand;
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
4
|
Endo M, Yazawa S, Sano R, Yokobori T, Shirabe K, Saeki H. α 1-Acid Glycoprotein with Highly Fucosylated Glycans as a Potential Diagnostic Marker for Early Detection of Hepatobiliary and Pancreatic Cancers. Diagnostics (Basel) 2024; 15:40. [PMID: 39795568 PMCID: PMC11720321 DOI: 10.3390/diagnostics15010040] [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: 11/22/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Previously, we reported elevated levels of fucosylated α1-acid glycoprotein (fAGP) in plasma samples from patients with diverse types of cancers. Accordingly, fAGP was assumed to be a potential biomarker for the early detection of cancers. Methods: The fAGP level was retrospectively measured in preoperative plasma samples from 213 patients with either hepatic, biliary tract, or pancreatic cancer and was analyzed together with levels of six existing tumor markers determined as reference standards. Results: When the cutoff value was set at 25.45 U/μg, elevated levels of fAGP were significantly observed in cancer patients. The sensitivity, specificity, and accuracy for the detection of malignancy in these diseases were determined to be 70.79, 51.72, and 68.12, respectively. In contrast, all the tumor markers exhibited low sensitivity and accuracy, even though they commonly had extremely high (≥80%) specificity. Further, a significant number of patients in both early and advanced clinical stages were found to be false negative in these tumor makers but were found to be positive in the fAGP level. A dramatic improvement in the diagnosis by tumor markers in such patients with all clinical stages was found by the determination of the fAGP level. This indicated that fAGP could serve to correct false-negative diagnosis with tumor markers. Conclusions: It is believed that fAGP could be a relevant, unique, and highly sensitive biomarker for early diagnosis of hepatobiliary and pancreatic cancers.
Collapse
Affiliation(s)
- Mizuki Endo
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan; (M.E.); (K.S.); (H.S.)
| | - Shin Yazawa
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan; (M.E.); (K.S.); (H.S.)
| | - Rie Sano
- Department of Legal Medicine, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan;
| | - Takehiko Yokobori
- Department of Innovative Cancer Immunotherapy, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan;
| | - Ken Shirabe
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan; (M.E.); (K.S.); (H.S.)
| | - Hiroshi Saeki
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan; (M.E.); (K.S.); (H.S.)
| |
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 2024. [PMID: 39714229 DOI: 10.1039/d4mo00187g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
Wang Z, Shi Y, Xiong G, Han M, Chen X. The prognostic impact of preoperative CA19-9 on resectable cholangiocarcinoma: a comprehensive systematic review and meta-analysis. Discov Oncol 2024; 15:773. [PMID: 39692970 DOI: 10.1007/s12672-024-01683-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND This study aimed to determine the prognostic survival impact of preoperative carbohydrate CA19-9 levels in resectable cholangiocarcinoma. METHODS PubMed, Web of Science, Cochrane, and Embase databases were searched for articles published through April 28, 2023. The relationship between preoperative serum carbohydrate antigen 19-9 and the prognosis of patients with resectable cholangiocarcinoma was analyzed. Heterogeneity between studies was assessed by means of the I2 statistic. We also performed subgroup analyses based on anatomical site of the tumor, geographic region, time of occurrence, and different levels of CA19-9. A random effects model was performed to express effect sizes as Hazard Ratio (HR) with 95% confidence intervals (CIs). RESULTS A total of 60 original studies were eligible for inclusion, with a total of 15,031 patients with all sites, including 9014 males. The overall Hazard Ratio for all studies was 1.90 (95% CI 1.74-2.07, p < 0.001, Z = 14.59). Publication bias was suggested by the Begg's test (p = 0.014 < 0.05), and the overall HR was 1.66 (95% CI 1.53-1.80, p < 0.001, Z = 12.027) after the trim-and-filling method. Subgroup analyses showed that intrahepatic cholangiocarcinoma (HR = 2.00, 95% CI 1.79-2.23), extrahepatic cholangiocarcinoma (HR = 1.65, 95% CI 1.49-1.82), hilar cholangiocarcinoma (HR = 1. 82, 95% CI 1.60-2.07), and distal cholangiocarcinoma (HR = 1.66, 95% CI 1.27-2.15) were predicted to be linked with prognosis. Elevated CA19-9 levels were associated with an increased risk of death. CONCLUSIONS This meta-analysis showed that elevated CA19-9 levels were correlated with a poor prognosis in cholangiocarcinoma. In future, more distal and hilar cholangiocarcinoma should be included in statistical studies to improve the accuracy of our conclusions.
Collapse
Affiliation(s)
- Zhicong Wang
- Department of Hepatobiliary Surgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
| | - Yabo Shi
- Department of General Surgery, Yellow River Sanmenxia Hospital, Sanmenxia, 472000, China
| | - Ganwei Xiong
- The First People's Hospital of Xiushui County, Jiujiang, 332400, China
| | - Mengxi Han
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xiaoliang Chen
- Department of Hepatobiliary Surgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China.
| |
Collapse
|
7
|
Facciorusso A, Crinò SF, Gkolfakis P, Spadaccini M, Arvanitakis M, Beyna T, Bronswijk M, Dhar J, Ellrichmann M, Gincul R, Hritz I, Kylänpää L, Martinez-Moreno B, Pezzullo M, Rimbaş M, Samanta J, van Wanrooij RLJ, Webster G, Triantafyllou K. Diagnostic work-up of bile duct strictures: European Society of Gastrointestinal Endoscopy (ESGE) Guideline. Endoscopy 2024. [PMID: 39689874 DOI: 10.1055/a-2481-7048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
1: ESGE recommends the combination of endoscopic ultrasound-guided tissue acquisition (EUS-TA) and endoscopic retrograde cholangiopancreatography (ERCP)-based tissue acquisition as the preferred diagnostic approach for tissue acquisition in patients with jaundice and distal extrahepatic biliary stricture in the absence of a pancreatic mass. 2: ESGE suggests that brushing cytology should be completed along with fluoroscopy-guided biopsies, wherever technically feasible, in patients with perihilar biliary strictures. 3: ESGE suggests EUS-TA for perihilar strictures when ERCP-based modalities yield insufficient results, provided that curative resection is not feasible and/or when cross-sectional imaging has shown accessible extraluminal disease. 4: ESGE suggests using standard ERCP diagnostic modalities at index ERCP. In the case of indeterminate biliary strictures, ESGE suggests cholangioscopy-guided biopsies, in addition to standard ERCP diagnostic modalities. Additional intraductal biliary imaging modalities can be selectively used, based on clinical context, local expertise, and resource availability.
Collapse
Affiliation(s)
- Antonio Facciorusso
- Experimental Medicine, Università del Salento, Lecce, Italy
- Clinical Effectiveness Research Group, Institute of Health and Society, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Paraskevas Gkolfakis
- Gastroenterology, "Konstantopoulio-Patision" General Hospital of Nea Ionia, Athens, Greece
| | | | - Marianna Arvanitakis
- Gastroenterology, Digestive Oncology and Hepatopancreatology, HUB Hôpital Erasme, Brussels, Belgium
| | - Torsten Beyna
- Internal Medicine, Evangelisches Krankenhaus Düsseldorf, Düsseldorf, Germany
| | - Michiel Bronswijk
- Gastroenterology and Hepatology, Imelda Hospital, Bonheiden, Belgium
- Gastroenterology and Hepatology, KU Leuven University Hospitals Leuven, Leuven, Belgium
| | | | - Mark Ellrichmann
- Interdisciplinary Endoscopy, Medical Department I, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Rodica Gincul
- Gastroenterology, Jean Mermoz Private Hospital, Lyon, France
| | - Istvan Hritz
- Centre for Therapeutic Endoscopy, Semmelweis University, Budapest, Hungary
| | - Leena Kylänpää
- Surgery, Helsinki Univeristy Central Hospital, Helsinki, Finland
| | | | | | - Mihai Rimbaş
- Gastroenterology, Colentina Clinical Hospital, Bucharest, Romania
- Internal Medicine Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Roy L J van Wanrooij
- Gastroenterology and Hepatology, Amsterdam UMC Locatie VUmc, Amsterdam, Netherlands
| | - George Webster
- Pancreatobiliary Medicine Unit, University College London, London, United Kingdom of Great Britain and Northern Ireland
| | - Konstantinos Triantafyllou
- Hepatogastroenterology Unit, 2nd Department of Internal Medicine, Propaedeutic, Medical School, National and Kapodistrian University of Athens, "Attikon" University General Hospital, Athens, Greece
| |
Collapse
|
8
|
Nakisa A, Sempere LF, Chen X, Qu LT, Woldring D, Crawford HC, Huang X. Tumor-Associated Carbohydrate Antigen 19-9 (CA 19-9), a Promising Target for Antibody-Based Detection, Diagnosis, and Immunotherapy of Cancer. ChemMedChem 2024; 19:e202400491. [PMID: 39230966 PMCID: PMC11648843 DOI: 10.1002/cmdc.202400491] [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: 06/27/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/06/2024]
Abstract
Carbohydrate antigen 19-9 (CA 19-9) also known as sialyl Lewis A is a tetrasaccharide overexpressed on a wide range of cancerous cells. CA 19-9 has been detected at elevated levels in sera of patients with various types of malignancies, most prominently pancreatic ductal adenocarcinoma. After its identification in 1979, multiple studies have highlighted the significant roles of CA 19-9 in cancer progression, including facilitating extravasation and eventually metastases, proliferation of cancer cells, and suppression of the immune system. Therefore, CA 19-9 has been considered an attractive target for cancer diagnosis, prognosis, and therapy. This review discusses the synthesis of CA 19-9 antigen, elicitation of antibodies through vaccination, development of anti-CA 19-9 monoclonal antibodies, and their applications as imaging tracers and therapeutics for a variety of CA 19-9-positive cancer.
Collapse
Affiliation(s)
- Athar Nakisa
- Department of ChemistryMichigan State UniversityEast Lansing, Michigan48824United States
- Institute for Quantitative Health Science and EngineeringMichigan State UniversityEast Lansing, Michigan48824United States
| | - Lorenzo F. Sempere
- Precision Health Program and Department of RadiologyMichigan State UniversityEast Lansing, Michigan48824United States
| | - Xi Chen
- Department of ChemistryUniversity of CaliforniaDavis, California95616USA
| | - Linda T. Qu
- Department of SurgeryMichigan State UniversityEast Lansing, Michigan48824United States
| | - Daniel Woldring
- Institute for Quantitative Health Science and EngineeringMichigan State UniversityEast Lansing, Michigan48824United States
- Department of Chemical Engineering and Materials ScienceMichigan State UniversityEast Lansing, Michigan48824United States
| | - Howard C. Crawford
- Department of SurgeryHenry Ford Health SystemDetroit, Michigan48202United States
- Department of Pharmacology and ToxicologyMichigan State UniversityEast Lansing, Michigan48824United States
| | - Xuefei Huang
- Department of ChemistryMichigan State UniversityEast Lansing, Michigan48824United States
- Institute for Quantitative Health Science and EngineeringMichigan State UniversityEast Lansing, Michigan48824United States
- Department of Biomedical EngineeringMichigan State UniversityEast Lansing, Michigan48824United States
| |
Collapse
|
9
|
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
|
10
|
Shen H, Zuo F. Prognostic role of systemic inflammation response index (SIRI) in patients with pancreatic cancer: a meta-analysis. Front Oncol 2024; 14:1465279. [PMID: 39723376 PMCID: PMC11668680 DOI: 10.3389/fonc.2024.1465279] [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: 07/16/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Background The significance of the systemic inflammation response index (SIRI) in predicting the prognosis of patients with pancreatic cancer (PC) has been extensively explored; however, findings remain controversial. As such, this meta-analysis was performed to more precisely determine the utility of the SIRI in predicting PC prognosis. Methods A comprehensive literature search of the PubMed, Web of Science, Embase, and Cochrane Library databases for relevant studies, published up to June 25, 2024, was performed. The primary and secondary endpoints were overall survival (OS) and progression-free survival (PFS), respectively. The prognostic utility of the SIRI in predicting PC prognosis was estimated by calculating pooled hazard ratios (HRs) and corresponding 95% confidence intervals (CIs). Results Seven studies comprising 1160 patients were included in the present meta-analysis. Pooled findings revealed that elevated SIRI was as a prominent prognostic marker of OS (HR 2.40 [95% CI 1.88-3.05]; p<0.001) and PFS (HR 1.95 [95% CI 1.19-3.21]; p=0.008) in patients diagnosed with PC. According to subgroup analysis, the SIRI remained an outstanding prognostic marker for OS, irrespective of region, sample size, study center, study design, TNM stage, cancer type, cut-off value, treatment, or survival analysis type (all p<0.05). Moreover, based on subgroup analysis, the SIRI demonstrated significant utility in predicting PFS, irrespective of region and threshold value (p<0.05). Conclusion Results of the present meta-analysis revealed that an increased SIRI significantly predicted OS and PFS in patients diagnosed with PC. Considering its cost-effectiveness and availability, the SIRI may be a promising biomarker for predicting prognosis in patients with PC.
Collapse
Affiliation(s)
- Huifen Shen
- Department of Neurology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Fei Zuo
- Department of Gastroenterology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, China
| |
Collapse
|
11
|
Maly M, Vanwalleghem L, Van Den Eeckhaut A, De Wilde V. Cancer of unknown primary and BRAF V600E meeting the BEACON combination: A case report. Mol Clin Oncol 2024; 21:88. [PMID: 39391046 PMCID: PMC11462391 DOI: 10.3892/mco.2024.2786] [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: 01/27/2024] [Accepted: 08/29/2024] [Indexed: 10/12/2024] Open
Abstract
The diagnostic work-up of cancer of unknown primary (CUP) is a challenging task; in addition, only a little data on BRAF targeting in CUP are currently available. Traditionally, the identification of favourable and unfavourable CUP subsets directs the choice of treatment. The present article reports the case of a 50-year-old male patient presenting with a BRAF-mutated CUP, a rare and generally unfavourable subset. Based on imaging, immunohistochemistry and a high value of carbohydrate antigen 19-9, an upper gastrointestinal profile was initially presumed. After disease progression on treatment with a first-line platinum-based doublet chemotherapy, a significant response was documented after treatment with the BEACON combination. The present case report highlighted the paradigm shift in diagnosis and treatment of CUP from a histology-based approach to molecular profiling with the introduction of precision medicine.
Collapse
Affiliation(s)
- Marlies Maly
- Department of Gastroenterology and Hepatology, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Gastroenterology and Hepatology, AZ Sint-Jan, 8000 Bruges, Belgium
| | | | | | - Vincent De Wilde
- Department of Gastroenterology and Hepatology, AZ Sint-Jan, 8000 Bruges, Belgium
| |
Collapse
|
12
|
Wang H, Ding Y, Zhao S, Li K, Li D. Establishment and validation of a nomogram model for early diagnosis of gastric cancer: a large-scale cohort study. Front Oncol 2024; 14:1463480. [PMID: 39678515 PMCID: PMC11638037 DOI: 10.3389/fonc.2024.1463480] [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: 07/12/2024] [Accepted: 11/12/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose Identifying high-risk populations and diagnosing gastric cancer (GC) early remains challenging. This study aimed to establish and verify a nomogram model for the early diagnosis of GC based on conventional laboratory indicators. Methods We performed a retrospective analysis of the clinical data of 2,770 individuals with first diagnosis of GC and 1,513 patients with benign gastric disease from January 2018 to December 2022. The cases were divided into the training set and validation set randomly, with a ratio of 7:3. Variable screening was performed by least absolute shrinkage and selection operator (LASSO) and logistic regression analysis. A nomogram was constructed in the training set to assist in the early diagnosis of GC. Results There were 4283 patients included in the study, with 2998 patients assigned in the training set and 1285 patients in the validation set. Through LASSO regression and logistic regression analysis, independent variables associated with GC were identified, including CEA, CA199, LYM, HGB, MCH, MCHC, PLT, ALB, TG, HDL, and AFR. The nomogram model was constructed using the above 11 independent indicators. The AUC was 0.803 for the training set and 0.797 for the validation set, indicating that the model showed high clinical diagnostic efficacy. The calibration curves and decision curve analysis (DCA) of the nomogram presented good calibration and clinical application ability. Conclusion Based on the analysis of large sample size, we constructed a nomogram model with 11 routine laboratory indicators, which showed good discrimination ability and calibration.
Collapse
Affiliation(s)
- Haiyu Wang
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yumin Ding
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Shujing Zhao
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Kaixu Li
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Dehong Li
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
13
|
Ma D, Wei P, Liu H, Hao J, Chen Z, Chu Y, Li Z, Shi W, Yuan Z, Cheng Q, Gao J, Zhu J, Li Z. Multi-omics-driven discovery of invasive patterns and treatment strategies in CA19-9 positive intrahepatic cholangiocarcinoma. J Transl Med 2024; 22:1031. [PMID: 39548460 PMCID: PMC11568536 DOI: 10.1186/s12967-024-05854-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (ICC) is a malignant tumor with a poor prognosis, predominantly CA19-9 positive. High CA19-9 levels correlate with increased aggressiveness and worse outcomes. This study employs multi-omics analysis to reveal molecular features and identify therapeutic targets of CA19-9 positive ICC, aiming to support individualized treatment. METHODS Data from seven clinical cohorts, two whole-exome sequencing cohorts, six RNA sequencing/microarray cohorts, one proteomic cohort, 20 single-cell RNA sequencing samples, and one spatial transcriptome sample were analyzed. Key findings were validated on tissue microarrays from 52 ICC samples. RESULTS CA19-9 positive ICC exhibited poorer OS (median 24.1 v.s. 51.5 months) and RFS (median 11.7 v.s. 28.2 months) compared to negative group (all P < 0.05). Genomic analysis revealed a higher KRAS mutation frequency in the positive group and a greater prevalence of IDH1/2 mutations in the negative group (all P < 0.05). Transcriptomic analysis indicated upregulated glycolysis pathways in CA19-9 positive ICC. Single-cell analysis identified specific glycolysis-related cell subclusters associated with poor prognosis, including Epi_SLC2A1, CAF_VEGFA, and Mph_SPP1. Higher hypoxia in the CA19-9 positive group led to metabolic reprogramming and promoted these cells' formation. These cells formed interactive communities promoting epithelial-mesenchymal transition (EMT) and angiogenesis. Drug sensitivity analysis identified six potential therapeutic drugs. CONCLUSIONS This study systematically elucidated the clinical, genomic, transcriptomic, and immune features of CA19-9 positive ICC. It reveals glycolysis-associated cellular communities and their cancer-promoting mechanisms, enhancing our understanding of ICC and laying the groundwork for individualized therapeutic strategies.
Collapse
Affiliation(s)
- Delin Ma
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of HCC and Liver Cirrhosis, Peking University People's Hospital, Beijing, China
- Peking University Center of Liver Cancer Diagnosis and Treatment, Peking University People's Hospital, Beijing, China
- Peking University Institute of Organ Transplantation, Peking University People's Hospital, Beijing, China
| | - Pengcheng Wei
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of HCC and Liver Cirrhosis, Peking University People's Hospital, Beijing, China
- Peking University Center of Liver Cancer Diagnosis and Treatment, Peking University People's Hospital, Beijing, China
- Peking University Institute of Organ Transplantation, Peking University People's Hospital, Beijing, China
| | - Hengkang Liu
- Peking University-Yunnan Baiyao International Medical Research Center, Beijing, 100191, China
| | - Jialing Hao
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of HCC and Liver Cirrhosis, Peking University People's Hospital, Beijing, China
- Peking University Center of Liver Cancer Diagnosis and Treatment, Peking University People's Hospital, Beijing, China
- Peking University Institute of Organ Transplantation, Peking University People's Hospital, Beijing, China
| | - Zhuomiaoyu Chen
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of HCC and Liver Cirrhosis, Peking University People's Hospital, Beijing, China
- Peking University Center of Liver Cancer Diagnosis and Treatment, Peking University People's Hospital, Beijing, China
- Peking University Institute of Organ Transplantation, Peking University People's Hospital, Beijing, China
| | - Yingming Chu
- Peking University First Hospital, Beijing, 100191, China
| | - Zuyin Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of HCC and Liver Cirrhosis, Peking University People's Hospital, Beijing, China
- Peking University Center of Liver Cancer Diagnosis and Treatment, Peking University People's Hospital, Beijing, China
- Peking University Institute of Organ Transplantation, Peking University People's Hospital, Beijing, China
| | - Wenzai Shi
- Department of Hepatobiliary Surgery, Peking University International Hospital, Life Park Road No.1 Life Science Park of Zhong Guancun, Chang Ping District, Beijing, 102206, China
| | - Zhigao Yuan
- Department of General Surgery, Civil Aviation General Hospital, Beijing, 100123, China
| | - Qian Cheng
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of HCC and Liver Cirrhosis, Peking University People's Hospital, Beijing, China
- Peking University Center of Liver Cancer Diagnosis and Treatment, Peking University People's Hospital, Beijing, China
- Peking University Institute of Organ Transplantation, Peking University People's Hospital, Beijing, China
| | - Jie Gao
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of HCC and Liver Cirrhosis, Peking University People's Hospital, Beijing, China
- Peking University Center of Liver Cancer Diagnosis and Treatment, Peking University People's Hospital, Beijing, China
- Peking University Institute of Organ Transplantation, Peking University People's Hospital, Beijing, China
| | - Jiye Zhu
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China.
- Beijing Key Laboratory of HCC and Liver Cirrhosis, Peking University People's Hospital, Beijing, China.
- Peking University Center of Liver Cancer Diagnosis and Treatment, Peking University People's Hospital, Beijing, China.
- Peking University Institute of Organ Transplantation, Peking University People's Hospital, Beijing, China.
| | - Zhao Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China.
- Beijing Key Laboratory of HCC and Liver Cirrhosis, Peking University People's Hospital, Beijing, China.
- Peking University Center of Liver Cancer Diagnosis and Treatment, Peking University People's Hospital, Beijing, China.
- Peking University Institute of Organ Transplantation, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
14
|
Bates M, Mohamed BM, Lewis F, O'Toole S, O'Leary JJ. Biomarkers in high grade serous ovarian cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189224. [PMID: 39581234 DOI: 10.1016/j.bbcan.2024.189224] [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: 01/28/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024]
Abstract
High-grade serous ovarian cancer (HGSC) is the most common subtype of ovarian cancer. HGSC patients typically present with advanced disease, which is often resistant to chemotherapy and recurs despite initial responses to therapy, resulting in the poor prognosis associated with this disease. There is a need to utilise biomarkers to manage the various aspects of HGSC patient care. In this review we discuss the current state of biomarkers in HGSC, focusing on the various available immunohistochemical (IHC) and blood-based biomarkers, which have been examined for their diagnostic, prognostic and theranostic potential in HGSC. These include various routine clinical IHC biomarkers such as p53, WT1, keratins, PAX8, Ki67 and p16 and clinical blood-borne markers and algorithms such as CA125, HE4, ROMA, RMI, ROCA, and others. We also discuss various components of the liquid biopsy as well as a number of novel IHC biomarkers and non-routine blood-borne biomarkers, which have been examined in various ovarian cancer studies. We also discuss the future of ovarian cancer biomarker research and highlight some of the challenges currently facing the field.
Collapse
Affiliation(s)
- Mark Bates
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland.
| | - Bashir M Mohamed
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Faye Lewis
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Sharon O'Toole
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin, Ireland
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin, Ireland
| |
Collapse
|
15
|
Shahbazlou SV, Vandghanooni S, Dabirmanesh B, Eskandani M, Hasannia S. Recent advances in surface plasmon resonance for the detection of ovarian cancer biomarkers: a thorough review. Mikrochim Acta 2024; 191:659. [PMID: 39382786 DOI: 10.1007/s00604-024-06740-3] [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: 04/09/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024]
Abstract
Early detection of ovarian cancer (OC) is crucial for effective management and treatment, as well as reducing mortality rates. However, the current diagnostic methods for OC are time-consuming and have low accuracy. Surface plasmon resonance (SPR) biosensors offer a promising alternative to conventional techniques, as they enable rapid and less invasive screening of various circulating indicators. These biosensors are widely used for biomolecular interaction analysis and detecting tumor markers, and they are currently being investigated as a rapid diagnostic tool for early-stage cancer detection. Our main focus is on the fundamental concepts and performance characteristics of SPR biosensors. We also discuss the latest advancements in SPR biosensors that enhance their sensitivity and enable high-throughput quantification of OC biomarkers, including CA125, HE4, CEA, and CA19-9. Finally, we address the future challenges that need to be overcome to advance SPR biosensors from research to clinical applications. The ultimate goal is to facilitate the translation of SPR biosensors into routine clinical practice for the early detection and management of OC.
Collapse
Affiliation(s)
- Shahnam Valizadeh Shahbazlou
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Research Center for Pharmaceutical Nanotechnology (RCPN), Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology (RCPN), Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Sadegh Hasannia
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
16
|
Wang W, Chen G, Zhang W, Zhang X, Huang M, Li C, Wang L, Lu Z, Xia J. The crucial prognostic signaling pathways of pancreatic ductal adenocarcinoma were identified by single-cell and bulk RNA sequencing data. Hum Genet 2024; 143:1109-1129. [PMID: 38526745 PMCID: PMC11485037 DOI: 10.1007/s00439-024-02663-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 02/24/2024] [Indexed: 03/27/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a malignant tumor with poor prognosis and high mortality. Although a large number of studies have explored its potential prognostic markers using traditional RNA sequencing (RNA-Seq) data, they have not achieved good prediction effect. In order to explore the possible prognostic signaling pathways leading to the difference in prognosis, we identified differentially expressed genes from one scRNA-seq cohort and four GEO cohorts, respectively. Then Cox and Lasso regression analysis showed that 12 genes were independent prognostic factors for PDAC. AUC and calibration curve analysis showed that the prognostic model had good discrimination and calibration. Compared with the low-risk group, the high-risk group had a higher proportion of gene mutations than the low-risk group. Immune infiltration analysis revealed differences in macrophages and monocytes between the two groups. Prognosis related genes were mainly distributed in fibroblasts, macrophages and type 2 ducts. The results of cell communication analysis showed that there was a strong communication between cancer-associated fibroblasts (CAF) and type 2 ductal cells, and collagen formation was the main interaction pathway.
Collapse
Affiliation(s)
- Wenwen Wang
- Department of Health Statistics, School of Military Preventive Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China.
| | - Guo Chen
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi Province, China
| | - Wenli Zhang
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi Province, China
| | - Xihua Zhang
- Department of Health Statistics, School of Military Preventive Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Sp
|
|