1
|
Jamal MH, Porel P, Aran KR. Emerging biomarkers for pancreatic cancer: from early detection to personalized therapy. Clin Transl Oncol 2025:10.1007/s12094-025-03947-5. [PMID: 40348906 DOI: 10.1007/s12094-025-03947-5] [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: 02/26/2025] [Accepted: 04/26/2025] [Indexed: 05/14/2025]
Abstract
Pancreatic cancer (PC) remains one of the most lethal malignancies, primarily due to its poor prognosis and late diagnosis. Biomarkers are essential in enhancing diagnostic accuracy, prognostic assessments, and therapeutic strategies, thereby addressing these challenges. Conventional biomarkers, such as CA 19-9, are widely used for monitoring disease progression but have limitations in early detection and specificity, necessitating complementary markers like CEA and MUC1. Emerging genetic biomarkers, including KRAS mutations and TP53 alterations, offer critical insights into tumorigenesis and serve as valuable diagnostic, prognostic, and therapeutic targets. Epigenetic biomarkers, such as DNA methylation and histone modifications, provide additional molecular layers, with aberrant methylation patterns and dysregulated histone modifications influencing tumor aggressiveness and therapy resistance. RNA-based biomarkers, particularly microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), play pivotal roles in regulating tumor biology and offer significant diagnostic and therapeutic potential. Protein-based biomarkers, including glycoproteins and cytokines, alongside liquid biopsy components like circulating tumor DNA (ctDNA), exosomes, and circulating tumor cells (CTCs), facilitate real-time disease monitoring and early detection. Personalized therapy is increasingly guided by these biomarkers, which predict responses to chemotherapy and immunotherapy. Despite challenges in biomarker validation and clinical implementation, advancements in multi-omics, artificial intelligence, and collaborative research hold promise for improving patient outcomes and survival rates in PC.
Collapse
Affiliation(s)
- Mohd Haris Jamal
- Department of Pharmacy Practice, ISF College of Pharmacy, GT Road, Moga, Punjab, 142001, India
| | - Pratyush Porel
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Moga, Punjab, 142001, India
| | - Khadga Raj Aran
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Moga, Punjab, 142001, India.
| |
Collapse
|
2
|
Xiao Y, Sun S, Zheng N, Zhao J, Li X, Xu J, Li H, Du C, Zeng L, Zhang J, Yin X, Huang Y, Yang X, Yuan F, Jia X, Li B, Li B. Development of PDAC diagnosis and prognosis evaluation models based on machine learning. BMC Cancer 2025; 25:512. [PMID: 40114140 PMCID: PMC11924714 DOI: 10.1186/s12885-025-13929-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is difficult to detect early and highly aggressive, often leading to poor patient prognosis. Existing serum biomarkers like CA19-9 are limited in early diagnosis, failing to meet clinical needs. Machine learning (ML)/deep learning (DL) technologies have shown great potential in biomedicine. This study aims to establish PDAC differential diagnosis and prognosis assessment models using ML combined with serum biomarkers for early diagnosis, risk stratification, and personalized treatment recommendations, improving early diagnosis rates and patient survival. METHODS The study included serum biomarker data and prognosis information from 117 PDAC patients. ML models (Random Forest (RF), Neural Network (NNET), Support Vector Machine (SVM), and Gradient Boosting Machine (GBM)) were used for differential diagnosis, evaluated by accuracy, Kappa test, ROC curve, sensitivity, and specificity. COX proportional hazards model and DeepSurv DL model predicted survival risk, compared by C-index and Log-rank test. Based on DeepSurv's risk predictions, personalized treatment recommendations were made and their effectiveness assessed. RESULTS Effective PDAC diagnosis and prognosis models were built using ML. The validation set data shows that the accuracy of the RF, NNET, SVM, and GBM models are 84.21%, 84.21%, 76.97%, and 83.55%; the sensitivity are 91.26%, 90.29%, 89.32%, and 88.35%; and the specificity are 69.39%, 71.43%, 51.02%, and 73.47%. The Kappa values are 0.6266, 0.6307, 0.4336, and 0.6215; and the AUC are 0.889, 0.8488, 0.8488, and 0.8704, respectively. BCAT1, AMY, and CA12-5 were selected as modeling parameters for the prognosis model using COX regression. DeepSurv outperformed the COX model on both training and validation sets, with C-indexes of 0.738 and 0.724, respectively. The Kaplan-Meier survival curves indicate that personalized treatment recommendations based on DeepSurv can help patients achieve survival benefits. CONCLUSION This study built efficient PDAC diagnosis and prognosis models using ML, improving early diagnosis rates and prognosis accuracy. The DeepSurv model excelled in prognosis prediction and successfully guided personalized treatment recommendations and supporting PDAC clinical management.
Collapse
Affiliation(s)
- Yingqi Xiao
- Department of Clinical Laboratory, Beijing Electric Power Teaching Hospital, Capital Medical University, Beijing, China
| | - Shixin Sun
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Naxin Zheng
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Jing Zhao
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Xiaohan Li
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Jianmin Xu
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Haolian Li
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Chenran Du
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Lijun Zeng
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Juling Zhang
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Xiuyun Yin
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Yuan Huang
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Xuemei Yang
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Fang Yuan
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Xingwang Jia
- Department of Clinical Laboratory, Beijing Electric Power Teaching Hospital, Capital Medical University, Beijing, China.
| | - Boan Li
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China.
| | - Bo Li
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
3
|
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
|
4
|
Jelski W, Mroczko J, Okrasinska S, Mroczko B. Diagnostic Utility of MicroRNAs in Pancreatic Cancers. Cancers (Basel) 2024; 16:3809. [PMID: 39594763 PMCID: PMC11593317 DOI: 10.3390/cancers16223809] [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: 10/08/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Pancreatic cancer (PC) is a serious malignant tumor with a high mortality rate, mainly due to late diagnosis and a lack of effective therapeutic interventions. The possibility of recognizing this cancer with reliable biomarkers using minimally invasive methods is of great importance for improving early detection, prognostic assessment, and targeted treatment methods. In recent years, small non-coding RNAs, especially microRNAs, have emerged as promising candidates for biomarkers of pancreatic cancer. Despite the methodological and analytical limitations of microRNA determination and a lack of available automated and standardized tests, a prospective systematic assessment of this new parameter's use in the detection of pancreatic cancer is warranted. This review provides general information on the diagnostic and prognostic utility of microRNAs, which appear to be well-established in many studies. In summary, microRNAs are promising non-invasive biomarkers of pancreatic cancer, offering potential opportunities for early detection, prognosis, and treatment monitoring. As research in this field progresses, microRNAs should become valuable tools in the fight against pancreatic cancer.
Collapse
Affiliation(s)
- Wojciech Jelski
- Department of Biochemical Diagnostics, Medical University, 15-276 Bialystok, Poland; (J.M.); (B.M.)
| | - Jan Mroczko
- Department of Biochemical Diagnostics, Medical University, 15-276 Bialystok, Poland; (J.M.); (B.M.)
| | - Sylwia Okrasinska
- Department of Biochemical Diagnostics, University Hospital, 15-268 Bialystok, Poland;
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, Medical University, 15-276 Bialystok, Poland; (J.M.); (B.M.)
- Department of Neurodegeneration Diagnostics, Medical University, 15-268 Bialystok, Poland
| |
Collapse
|
5
|
Oi H, Hozaka Y, Akahane T, Fukuda K, Idichi T, Tanoue K, Yamasaki Y, Kawasaki Y, Mataki Y, Kurahara H, Higashi M, Tanimoto A, Ohtsuka T. Genetic Assessment of Intraductal Papillary Mucinous Neoplasm for Predicting Concomitant Pancreatic Ductal Adenocarcinoma. Pancreas 2024; 53:e790-e795. [PMID: 38743932 DOI: 10.1097/mpa.0000000000002373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
OBJECTIVE The role of Krüppel-like transcription factor 4 ( KLF4 ) mutations in IPMNs with concomitant pancreatic ductal adenocarcinoma (PDAC) remains unclear. This study clarified the rate and effect of KLF4 mutations in IPMN with concomitant PDAC. MATERIALS AND METHODS DNA was extracted from 65 formalin-fixed and paraffin-embedded samples from 52 patients including 13 IPMNs with concomitant PDAC and 39 IPMNs alone. A comprehensive screening using next-generation sequencing and then targeted sequencing for KLF4 , GNAS , and KRAS mutations were performed. RESULTS In next-generation sequencing screening, KRAS mutations were observed in all samples except for one, GNAS mutation in 2 IPMNs with concomitant PDAC, and a KLF4 mutation in 1 IPMN with concomitant PDAC. Targeted sequence detected KLF4 mutations in 11 of the 52 IPMNs. Concomitant PDAC developed only in the nonintestinal, noninvasive, and branch-duct IPMNs, and KLF4 mutations were more frequent in this IPMN type than in the other type. For this IPMN type with KLF4 mutation, PDAC-prediction sensitivity, specificity, and accuracy were 63%, 82%, and 79%, respectively. CONCLUSION For selected IPMNs with nonintestinal, noninvasive, and branch-duct, genetic assessment might be helpful for predicting the possible development of concomitant PDAC, although a prospective validation study using a larger study population is needed.
Collapse
Affiliation(s)
| | | | - Toshiaki Akahane
- Pathology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | | | | | | | | | | | | | | | - Michiyo Higashi
- Pathology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Akihide Tanimoto
- Pathology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | | |
Collapse
|
6
|
Tavano F, Latiano A, Palmieri O, Gioffreda D, Latiano T, Gentile A, Tardio M, Latiano TP, Gentile M, Terracciano F, Perri F. Duodenal Fluid Analysis as a Rewarding Approach to Detect Low-Abundance Mutations in Biliopancreatic Cancers. Int J Mol Sci 2024; 25:8436. [PMID: 39126005 PMCID: PMC11312909 DOI: 10.3390/ijms25158436] [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: 06/20/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Diagnosis of biliopancreatic cancers by the available serum tumor markers, imaging, and histopathological tissue specimen examination remains a challenge. Circulating cell-free DNA derived from matched pairs of secretin-stimulated duodenal fluid (DF) and plasma from 10 patients with biliopancreatic diseases and 8 control subjects was analyzed using AmpliSeq™ HD technology for Ion Torrent Next-Generation Sequencing to evaluate the potential of liquid biopsy with DF in biliopancreatic cancers. The median cfDNA concentration was greater in DF-derived than in plasma-derived samples. A total of 13 variants were detected: 11 vs. 1 were exclusive for DF relative to the plasma source, and 1 was shared between the two body fluids. According to the four-tier systems, 10 clinical tier-I-II (76.9%), 1 tier-III (7.7%), and 2 tier-IV (15.4%) variants were identified. Notably, the 11 tier-I-III variants were exclusively found in DF-derived cfDNA from five patients with biliopancreatic cancers, and were detected in seven genes (KRAS, TP53, BRAF, CDKN2A, RNF43, GNAS, and PIK3CA); 82% of the tier-I-III variants had a low abundance, with a VAF < 6%. The mutational profiling of DF seems to be a reliable and promising tool for identifying cancer-associated alterations in malignant cancers of the biliopancreatic tract.
Collapse
Affiliation(s)
- Francesca Tavano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Anna Latiano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Orazio Palmieri
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Domenica Gioffreda
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Tiziana Latiano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Annamaria Gentile
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Matteo Tardio
- Department of Surgery, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Tiziana Pia Latiano
- Department of Oncology, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Marco Gentile
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Fulvia Terracciano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Francesco Perri
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| |
Collapse
|
7
|
Lee YS, Im J, Yang Y, Lee HJ, Lee MR, Woo SM, Park SJ, Kong SY, Kim JY, Hwang H, Kim YH. New Function Annotation of PROSER2 in Pancreatic Ductal Adenocarcinoma. J Proteome Res 2024; 23:905-915. [PMID: 38293943 PMCID: PMC10913870 DOI: 10.1021/acs.jproteome.3c00632] [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: 09/27/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 02/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis due to the absence of diagnostic markers and molecular targets. Here, we took an unconventional approach to identify new molecular targets for pancreatic cancer. We chose uncharacterized protein evidence level 1 without function annotation from extensive proteomic research on pancreatic cancer and focused on proline and serine-rich 2 (PROSER2), which ranked high in the cell membrane and cytoplasm. In our study using cell lines and patient-derived orthotopic xenograft cells, PROSER2 exhibited a higher expression in cells derived from primary tumors than in those from metastatic tissues. PROSER2 was localized in the cell membrane and cytosol by immunocytochemistry. PROSER2 overexpression significantly reduced the metastatic ability of cancer cells, whereas its suppression had the opposite effect. Proteomic analysis revealed that PROSER2 interacts with STK25 and PDCD10, and their binding was confirmed by immunoprecipitation and immunocytochemistry. STK25 knockdown enhanced metastasis by decreasing p-AMPK levels, whereas PROSER2-overexpressing cells increased the level of p-AMPK, indicating that PROSER2 suppresses invasion via the AMPK pathway by interacting with STK25. This is the first demonstration of the novel role of PROSER2 in antagonizing tumor progression via the STK25-AMPK pathway in PDAC. LC-MS/MS data are available at MassIVE (MSV000092953) and ProteomeXchange (PXD045646).
Collapse
Affiliation(s)
- Yu-Sun Lee
- Division
of Convergence Technology, Research Institute
of National Cancer Center, Goyang 10408, Republic
of Korea
- Department
of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic
of Korea
| | - Jieun Im
- Division
of Convergence Technology, Research Institute
of National Cancer Center, Goyang 10408, Republic
of Korea
| | - Yeji Yang
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Cheongju 28119, Republic
of Korea
- Critical
Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Hea Ji Lee
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Cheongju 28119, Republic
of Korea
| | - Mi Rim Lee
- Department
of Cancer Biomedical Science, National Cancer
Center Graduate School of Cancer Science and Policy, Goyang 10408, Republic of Korea
| | - Sang-Myung Woo
- Department
of Cancer Biomedical Science, National Cancer
Center Graduate School of Cancer Science and Policy, Goyang 10408, Republic of Korea
- Department
of Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic
of Korea
| | - Sang-Jae Park
- Department
of Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic
of Korea
- Department
of Surgical Oncology Branch, Research Institute
of National Cancer Center, Goyang 10408, Republic
of Korea
| | - Sun-Young Kong
- Department
of Cancer Biomedical Science, National Cancer
Center Graduate School of Cancer Science and Policy, Goyang 10408, Republic of Korea
- Department
of Targeted Therapy Branch, Research Institute
of National Cancer Center, Goyang 10408, Republic
of Korea
| | - Jin Young Kim
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Cheongju 28119, Republic
of Korea
- Critical
Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Heeyoun Hwang
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Cheongju 28119, Republic
of Korea
- Critical
Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Yun-Hee Kim
- Division
of Convergence Technology, Research Institute
of National Cancer Center, Goyang 10408, Republic
of Korea
- Department
of Cancer Biomedical Science, National Cancer
Center Graduate School of Cancer Science and Policy, Goyang 10408, Republic of Korea
| |
Collapse
|
8
|
Tharrun Daniel Paul L, Munuswamy-Ramanujam G, Kumar RCS, Ramachandran V, Gnanasampanthapandian D, Palaniyandi K. Recent advancement in molecular markers of pancreatic cancer. BIOMARKERS IN CANCER DETECTION AND MONITORING OF THERAPEUTICS 2024:121-149. [DOI: 10.1016/b978-0-323-95114-2.00025-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
9
|
Alexandrou E, Guneri D, Neidle S, Waller ZAE. QN-302 demonstrates opposing effects between i-motif and G-quadruplex DNA structures in the promoter of the S100P gene. Org Biomol Chem 2023; 22:55-58. [PMID: 37970888 PMCID: PMC10732280 DOI: 10.1039/d3ob01464a] [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: 09/12/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023]
Abstract
GC-rich sequences can fold into G-quadruplexes and i-motifs and are known to control gene expression in many organisms. The potent G-quadruplex experimental anticancer drug QN-302 down-regulates a number of cancer-related genes, in particular S100P. Here we show this ligand has strong opposing effects with i-motif DNA structures and is one of the most potent i-motif destabilising agents reported to date. QN-302 down-regulates the expression of numerous cancer-related genes by pan-quadruplex targeting. QN-302 exhibits exceptional combined synergistic effects compared to many other G-quadruplex and i-motif interacting compounds. This work further emphasises the importance of considering G-quadruplex and i-motif DNA structures as one dynamic system.
Collapse
Affiliation(s)
- Effrosyni Alexandrou
- School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK.
| | - Dilek Guneri
- School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK.
| | - Stephen Neidle
- School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK.
| | - Zoë A E Waller
- School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK.
| |
Collapse
|
10
|
Farahmand Y, Tehrany PM, Nazari A, Nava ZH, Alsaffar MF, Yazdani O, Adili A, Esbati R, Ghafouri K. A comprehensive survey into the role of exosomes in pancreatic cancer; from the origin of cancer to the progress and possibility of diagnosis and treatment. Pathol Res Pract 2023; 245:154465. [PMID: 37119731 DOI: 10.1016/j.prp.2023.154465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/09/2023] [Accepted: 04/12/2023] [Indexed: 05/01/2023]
Abstract
Pancreatic cancer is the fourth most common malignant tumor in the world, which has a high mortality rate due to high invasiveness, early metastases, lack of specific symptoms, and high invasiveness. Recent studies have shown that exosomes can be essential sources of biomarkers in pancreatic cancer. Over the past ten years, exosomes have been implicated in multiple trials to prevent the growth and metastasis of many cancers, including pancreatic cancer. Exosomes also play essential roles in immune evasion, invasion, metastasis, proliferation, apoptosis, drug resistance, and cancer stemness. Exosomes help cells communicate by carrying proteins and genetic material, such as non-coding RNAs, including mRNAs and microRNAs. This review examines the biological significance of exosomes in pancreatic cancer and their functions in tumor invasion, metastasis, treatment resistance, proliferation, stemness, and immune evasion. We also emphasize recent advances in our understanding of the main functions of exosomes in diagnosing and treating pancreatic cancer.
Collapse
Affiliation(s)
- Yalda Farahmand
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Pooya M Tehrany
- Faculty of Medicine, National University of Malaysia, Bani, Malaysia
| | - Ahmad Nazari
- Tehran University of Medical Sciences, Tehran, Iran
| | | | - Marwa Fadhil Alsaffar
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, 51001 Hillah, Babil, Iraq
| | - Omid Yazdani
- Department of Medicine, Shahid Beheshti University, Tehran, Iran
| | - Ali Adili
- Tabriz University of Medical Sciences, Tabriz, Iran
| | - Romina Esbati
- Department of Medicine, Shahid Beheshti University, Tehran, Iran.
| | - Kimia Ghafouri
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Wnuk J, Strzelczyk JK, Gisterek I. Clinical Value of Circulating miRNA in Diagnosis, Prognosis, Screening and Monitoring Therapy of Pancreatic Ductal Adenocarcinoma-A Review of the Literature. Int J Mol Sci 2023; 24:ijms24065113. [PMID: 36982210 PMCID: PMC10049684 DOI: 10.3390/ijms24065113] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/28/2023] [Accepted: 03/04/2023] [Indexed: 03/30/2023] Open
Abstract
Pancreatic cancer (PC) is considered to be the seventh most common cause of cancer-related deaths. The number of deaths caused by PC is estimated to increase in the future. An early diagnosis of PC is crucial for improving treatment outcomes. The most common histopathological subtype of PC is pancreatic ductal adenocarcinoma (PDAC). MicroRNAs (miRNAs)-which are endogenous non-coding RNAs involved in the posttranscriptional regulation of multiple gene expression-constitute useful diagnostic and prognostic biomarkers in various neoplasms, including PDAC. Circulating miRNAs detected in a patient's serum or plasma are drawing more and more attention. Hence, this review aims at evaluating the clinical value of circulating miRNA in the screening, diagnosis, prognosis and monitoring of pancreatic ductal adenocarcinoma therapy.
Collapse
Affiliation(s)
- Jakub Wnuk
- Department of Oncology and Radiotherapy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 35 Ceglana St., 40-515 Katowice, Poland
| | - Joanna Katarzyna Strzelczyk
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
| | - Iwona Gisterek
- Department of Oncology and Radiotherapy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 35 Ceglana St., 40-515 Katowice, Poland
| |
Collapse
|
12
|
Ahmed AA, Greenhalf W, Palmer DH, Williams N, Worthington J, Arshad T, Haider S, Alexandrou E, Guneri D, Waller ZAE, Neidle S. The Potent G-Quadruplex-Binding Compound QN-302 Downregulates S100P Gene Expression in Cells and in an In Vivo Model of Pancreatic Cancer. Molecules 2023; 28:molecules28062452. [PMID: 36985425 PMCID: PMC10051992 DOI: 10.3390/molecules28062452] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/30/2023] Open
Abstract
The naphthalene diimide compound QN-302, designed to bind to G-quadruplex DNA sequences within the promoter regions of cancer-related genes, has high anti-proliferative activity in pancreatic cancer cell lines and anti-tumor activity in several experimental models for the disease. We show here that QN-302 also causes downregulation of the expression of the S100P gene and the S100P protein in cells and in vivo. This protein is well established as being involved in key proliferation and motility pathways in several human cancers and has been identified as a potential biomarker in pancreatic cancer. The S100P gene contains 60 putative quadruplex-forming sequences, one of which is in the promoter region, 48 nucleotides upstream from the transcription start site. We report biophysical and molecular modeling studies showing that this sequence forms a highly stable G-quadruplex in vitro, which is further stabilized by QN-302. We also report transcriptome analyses showing that S100P expression is highly upregulated in tissues from human pancreatic cancer tumors, compared to normal pancreas material. The extent of upregulation is dependent on the degree of differentiation of tumor cells, with the most poorly differentiated, from more advanced disease, having the highest level of S100P expression. The experimental drug QN-302 is currently in pre-IND development (as of Q1 2023), and its ability to downregulate S100P protein expression supports a role for this protein as a marker of therapeutic response in pancreatic cancer. These results are also consistent with the hypothesis that the S100P promoter G-quadruplex is a potential therapeutic target in pancreatic cancer at the transcriptional level for QN-302.
Collapse
Affiliation(s)
- Ahmed A Ahmed
- The School of Pharmacy, University College London, London WC1N 1AX, UK
- Guy's Cancer Centre, Guy's Hospital, London SE1 9RT, UK
| | - William Greenhalf
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 7BE, UK
| | - Daniel H Palmer
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 7BE, UK
| | | | | | | | - Shozeb Haider
- The School of Pharmacy, University College London, London WC1N 1AX, UK
| | | | - Dilek Guneri
- The School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Zoe A E Waller
- The School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Stephen Neidle
- The School of Pharmacy, University College London, London WC1N 1AX, UK
| |
Collapse
|
13
|
Zhuang L, Huang C, Ning Z, Yang L, Zou W, Wang P, Cheng CS, Meng Z. Circulating tumor-associated autoantibodies as novel diagnostic biomarkers in pancreatic adenocarcinoma. Int J Cancer 2023; 152:1013-1024. [PMID: 36274627 DOI: 10.1002/ijc.34334] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 01/06/2023]
Abstract
To develop a superior diagnostic approach for pancreatic adenocarcinoma (PAAC), the present study prospectively included 338 PAAC patients, 294 normal healthy volunteers (NHV), 122 chronic pancreatitis (CP) patients and 100 patients with non-PAAC malignancies. In the identification phase, HuProt Human Proteome Microarray, comprising 21 065 proteins, was used to identify serum tumor-associated autoantibodies (TAAbs) candidates differentiating PAAC (n = 30) from NHV (n = 30). A PAAC-focused array containing 165 differentially expressed TAAbs identified was subsequently adopted in the validation phase (n = 712) for specificity and sensitivities. The multivariate TAAbs signature for differentiation PAAC from controls (NHV + CP) identified five candidates, namely the IgG-type TAAbs against CLDN17, KCNN3, SLAMF7, SLC22A11 and OR51F2. Multivariate logistic performance model of y = (22.893 × CA19-9 + 0.68 × CLDN17 - 4.012) showed a significant better diagnostic accuracy than that of CA19-9 and CLDN17 in differentiating PAAC from controls (NHV + CP) (AUC = 0.97, 0.92 and 0.82, respectively, P-value < .0001). We further tested the autoantigen level of CLDN17 by ELISA in 82 sera samples from PAAC (n = 42), CP (n = 24) and NHV (n = 16). Similarly, the model showed superior diagnostic performance than that of CA19-9 and CLDN17 (AUC = 0.93, 0.83 and 0.81, respectively, P-value < .0001) in differentiating PAAC from controls. In conclusion, our study is the first to characterize the circulating TAAbs signatures in PAAC. The results showed that CLDN17 combined with CA19-9 provided potentially clinical value and may serve as noninvasive novel biomarkers for PAAC diagnosis.
Collapse
Affiliation(s)
- Liping Zhuang
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Changjing Huang
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhouyu Ning
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lina Yang
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Shandong Province, China
| | - Wenbin Zou
- Department of Gastroenterology, Digestive Endoscopy Center, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Peng Wang
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chien-Shan Cheng
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiqiang Meng
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Bunduc S, Gede N, Váncsa S, Lillik V, Kiss S, Juhász MF, Erőss B, Szakács Z, Gheorghe C, Mikó A, Hegyi P. Exosomes as prognostic biomarkers in pancreatic ductal adenocarcinoma-a systematic review and meta-analysis. Transl Res 2022; 244:126-136. [PMID: 35066189 DOI: 10.1016/j.trsl.2022.01.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 01/09/2022] [Accepted: 01/13/2022] [Indexed: 01/06/2023]
Abstract
Extensive research is focused on the role of liquid biopsy in pancreatic cancer since reliable diagnostic and follow-up biomarkers represent an unmet need for this highly lethal malignancy. We performed a systematic review and meta-analysis on the prognostic value of exosomal biomarkers in pancreatic ductal adenocarcinoma (PDAC). MEDLINE, Embase, Scopus, Web of Science, and CENTRAL were systematically searched on the 18th of January, 2021 for studies reporting on the differences in overall (OS) and progression-free survival (PFS) in PDAC patients with positive vs negative exosomal biomarkers isolated from blood. The random-effects model estimated pooled multivariate-adjusted (AHR) and univariate hazard ratios (UHRs) with 95% confidence intervals (CIs). Eleven studies comprising 634 patients were eligible for meta-analysis. Detection of positive exosomal biomarkers indicated increased risk of mortality (UHR = 2.81, CI:1.31-6,00, I2 = 88.7%, P < 0.001), and progression (UHR = 3.33, CI: 2.33-4.77, I2 = 0, P = 0.879) across various disease stages. Positive exosomal biomarkers identified preoperatively revealed a higher risk of mortality in resectable stages (UHR = 5.55, CI: 3.24-9.49, I2 = 0, P = 0.898). The risk of mortality in unresectable stages was not significantly increased with positive exosomal biomarkers (UHR = 2.51, CI: 0.55-11.43, I2 = 90.3%, P < 0.001). Detectable exosomal micro ribonucleic acids were associated with a decreased OS (UHR = 4.08, CI: 2.16-7.69, I2 = 46.9%, P = 0.152) across various stages. Our results reflect the potential of exosomal biomarkers for prognosis evaluation in PDAC. The associated heterogeneity reflects the variability of study methods and need for their uniformization before transition to clinical use.
Collapse
Affiliation(s)
- Stefania Bunduc
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Szigeti ú;t 12, Hungary; Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; Fundeni Clinical Institute, 022328 Bucharest, Romania; Center for Translational Medicine, Semmelweis University, 1085 Budapest, Üllői út 26, Hungary; Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, 1085 Budapest, Baross út 22-24, Hungary
| | - Noémi Gede
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Szigeti ú;t 12, Hungary; János Szentágothai Research Center, University of Pécs, 7624 Pécs, Szigeti út 12, Hungary
| | - Szilárd Váncsa
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Szigeti ú;t 12, Hungary; János Szentágothai Research Center, University of Pécs, 7624 Pécs, Szigeti út 12, Hungary; Center for Translational Medicine, Semmelweis University, 1085 Budapest, Üllői út 26, Hungary; Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, 1085 Budapest, Baross út 22-24, Hungary
| | - Veronika Lillik
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Szigeti ú;t 12, Hungary
| | - Szabolcs Kiss
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Szigeti ú;t 12, Hungary; Doctoral School of Clinical Medicine, University of Szeged, 6720, Hungary
| | - Márk Félix Juhász
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Szigeti ú;t 12, Hungary; János Szentágothai Research Center, University of Pécs, 7624 Pécs, Szigeti út 12, Hungary; Center for Translational Medicine, Semmelweis University, 1085 Budapest, Üllői út 26, Hungary
| | - Bálint Erőss
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Szigeti ú;t 12, Hungary; János Szentágothai Research Center, University of Pécs, 7624 Pécs, Szigeti út 12, Hungary; Center for Translational Medicine, Semmelweis University, 1085 Budapest, Üllői út 26, Hungary; Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, 1085 Budapest, Baross út 22-24, Hungary
| | - Zsolt Szakács
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Szigeti ú;t 12, Hungary; First Department of Medicine, Medical School, University of Pécs, 7624 Pécs, Szigeti út 12 Hungary
| | - Cristian Gheorghe
- Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Alexandra Mikó
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Szigeti ú;t 12, Hungary; Department of Medical Genetics, Medical School, University of Pécs, 7623, Pécs, József Attila út 7
| | - Péter Hegyi
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Szigeti ú;t 12, Hungary; János Szentágothai Research Center, University of Pécs, 7624 Pécs, Szigeti út 12, Hungary; Center for Translational Medicine, Semmelweis University, 1085 Budapest, Üllői út 26, Hungary; Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, 1085 Budapest, Baross út 22-24, Hungary.
| |
Collapse
|
15
|
Vivekanandhan S, Madamsetty VS, Angom RS, Dutta SK, Wang E, Caulfield T, Pletnev AA, Upstill-Goddard R, Asmann YW, Chang D, Spaller MR, Mukhopadhyay D. Role of PLEXIND1/TGFβ Signaling Axis in Pancreatic Ductal Adenocarcinoma Progression Correlates with the Mutational Status of KRAS. Cancers (Basel) 2021; 13:cancers13164048. [PMID: 34439202 PMCID: PMC8393884 DOI: 10.3390/cancers13164048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/04/2021] [Accepted: 08/09/2021] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Pancreatic cancer is among the most lethal cancers. The expression of PLEXIND1, a receptor, is upregulated in many cancers (including pancreatic cancer). Traditionally, PLEXIND1 is known to be involved in neuron development and mediate semaphorin signaling. However, its role and signaling in cancer is not fully understood. In our study, we present a new mechanism through which PLEXIND1 mediates its roles in cancer. For the first time, we demonstrate that it can function as a transforming growth factor beta coreceptor and modulate SMAD3 signaling. Around 90% of pancreatic cancer patients have mutant KRAS. Our work suggests that PLEXIND1 functions differently in pancreatic cancer cell lines, and the difference correlates with KRAS mutational status. Additionally, we demonstrate a novel peptide based therapeutic approach to target PLEXIND1 in cancer cells. Our work is valuable to both neuroscience and cancer fields, as it demonstrates an association between two previously unrelated signaling pathways. Abstract PLEXIND1 is upregulated in several cancers, including pancreatic ductal adenocarcinoma (PDAC). It is an established mediator of semaphorin signaling, and neuropilins are its known coreceptors. Herein, we report data to support the proposal that PLEXIND1 acts as a transforming growth factor beta (TGFβ) coreceptor, modulating cell growth through SMAD3 signaling. Our findings demonstrate that PLEXIND1 plays a pro-tumorigenic role in PDAC cells with oncogenic KRAS (KRASmut). We show in KRASmut PDAC cell lines (PANC-1, AsPC-1,4535) PLEXIND1 downregulation results in decreased cell viability (in vitro) and reduced tumor growth (in vivo). Conversely, PLEXIND1 acts as a tumor suppressor in the PDAC cell line (BxPC-3) with wild-type KRAS (KRASwt), as its reduced expression results in higher cell viability (in-vitro) and tumor growth (in vivo). Additionally, we demonstrate that PLEXIND1-mediated interactions can be selectively disrupted using a peptide based on its C-terminal sequence (a PDZ domain-binding motif), an outcome that may possess significant therapeutic implications. To our knowledge, this is the first report showing that (1) PLEXIND1 acts as a TGFβ coreceptor and mediates SMAD3 signaling, and (2) differential roles of PLEXIND1 in PDAC cell lines correlate with KRASmut and KRASwt status.
Collapse
Affiliation(s)
- Sneha Vivekanandhan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Vijay S. Madamsetty
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Shamit Kumar Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Thomas Caulfield
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Alexandre A. Pletnev
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA; (A.A.P.); (M.R.S.)
| | - Rosanna Upstill-Goddard
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate Switchback Road, Glasgow G12 8QQ, UK; (R.U.-G.); (D.C.)
| | - Yan W. Asmann
- Health Sciences Research, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA;
| | - David Chang
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate Switchback Road, Glasgow G12 8QQ, UK; (R.U.-G.); (D.C.)
| | - Mark R. Spaller
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA; (A.A.P.); (M.R.S.)
- Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, Lebanon, NH 03756, USA
- Division of Natural and Applied Sciences, Duke Kunshan University, Kunshan 215316, China
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
- Correspondence:
| |
Collapse
|
16
|
Zhao Z, Zhao G, Yang S, Zhu S, Zhang S, Li P. The significance of exosomal RNAs in the development, diagnosis, and treatment of pancreatic cancer. Cancer Cell Int 2021; 21:364. [PMID: 34243775 PMCID: PMC8268510 DOI: 10.1186/s12935-021-02059-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/27/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are single-membrane, secreted organelles with a diameter of 30–200 nm, containing diverse bioactive constituents, including DNAs, RNAs, proteins, and lipids, with prominent molecular heterogeneity. Extensive studies indicate that exosomal RNAs (e.g., microRNAs, long non-coding RNAs, and circular RNAs) can interact with many types of cancers, associated with several hallmark features like tumor growth, metastasis, and resistance to therapy. Pancreatic cancer (PaCa) is among the most lethal cancers worldwide, emerging as the seventh foremost cause of cancer-related death in both sexes. Hence, revealing the specific pathogenesis and improving the clinical diagnosis and treatment process are urgently required. As the study of exosomes has become an active area of research, the functional connections between exosomes and PaCa have been deeply investigated. Among these, exosomal RNAs seem to play a significant role in the development, diagnosis, and treatment of PaCa. Exosomal RNAs delivery ultimately modulates the various features of PaCa, and many scholars have interpreted how exosomal RNAs contribute to the proliferation, angiogenesis, migration, invasion, metastasis, immune escape, and drug resistance in PaCa. Besides, recent studies emphasize that exosomal RNAs may serve as diagnostic and prognostic biomarkers or therapeutic targets for PaCa. In this review, we will introduce these recent insights focusing on the discoveries of the relationship between exosomal RNAs and PaCa, and the potentially diagnostic and therapeutic applications of exosomes in PaCa.
Collapse
Affiliation(s)
- Zheng Zhao
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong'an Road, Xicheng District, Beijing, 100050, China
| | - Guiping Zhao
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong'an Road, Xicheng District, Beijing, 100050, China.
| | - Shuyue Yang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong'an Road, Xicheng District, Beijing, 100050, China
| | - Shengtao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong'an Road, Xicheng District, Beijing, 100050, China
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong'an Road, Xicheng District, Beijing, 100050, China
| | - Peng Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong'an Road, Xicheng District, Beijing, 100050, China.
| |
Collapse
|
17
|
Mehta S, Bhimani N, Gill AJ, Samra JS, Sahni S, Mittal A. Serum Biomarker Panel for Diagnosis and Prognosis of Pancreatic Ductal Adenocarcinomas. Front Oncol 2021; 11:708963. [PMID: 34290990 PMCID: PMC8287202 DOI: 10.3389/fonc.2021.708963] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
Background Patients with pancreatic ductal adenocarcinoma (PDAC) have late diagnosis which results in poor prognosis. Currently, surgical resection is the only option for curative intent. Identifying high-risk features for patients with aggressive PDAC is essential for accurate diagnosis, prognostication, and personalised care due to the disease burden and risk of recurrence despite surgical resection. A panel of three biomarkers identified in tumour tissue (S100A4, Ca125 and Mesothelin) have shown an association with poor prognosis and overall survival. The diagnostic and prognostic value of the serum concentration of this particular biomarker panel for patients with PDAC has not been previously studied. Methods Retrospectively collected blood samples of PDAC patients (n =120) and healthy controls (n =80) were evaluated for the serum concentration of select biomarkers - S100A4, S100A2, Ca-125, Ca 19-9 and mesothelin. Statistical analyses were performed for diagnostic and prognostic correlation. Results A panel of four biomarkers (S100A2, S100A4, Ca-125 and Ca 19-9) achieved high diagnostic potential (AUROC 0.913). Three biomarkers (S100A4, Ca-125 and Ca 19-9) correlated with poor overall survival in a univariable model (p < 0.05). PDAC patients with abnormal levels of 2 or more biomarkers in their serum demonstrated significantly lower survival compared to patients with abnormal levels of one or less biomarker (p < 0.05). Conclusion and Impact The identified biomarker panels have shown the potential to diagnose PDAC patients and stratify patients based on their prognostic outcomes. If independently validated, this may lead to the development of a diagnostic and prognosticating blood test for PDAC.
Collapse
Affiliation(s)
- Shreya Mehta
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.,Kolling Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Nazim Bhimani
- Upper Gastro Intestinal (GI) Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, Sydney, NSW, Australia
| | - Anthony J Gill
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.,Kolling Institute of Medical Research, University of Sydney, Sydney, NSW, Australia.,Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia.,Australian Pancreatic Centre, Sydney, NSW, Australia.,NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Jaswinder S Samra
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.,Upper Gastro Intestinal (GI) Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, Sydney, NSW, Australia.,Australian Pancreatic Centre, Sydney, NSW, Australia
| | - Sumit Sahni
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.,Kolling Institute of Medical Research, University of Sydney, Sydney, NSW, Australia.,Australian Pancreatic Centre, Sydney, NSW, Australia
| | - Anubhav Mittal
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.,Upper Gastro Intestinal (GI) Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, Sydney, NSW, Australia.,Australian Pancreatic Centre, Sydney, NSW, Australia
| |
Collapse
|
18
|
Cui J, Yang H, Liu J, Chen D, Hu J, Zhang H, Wang Y, Han T, Mao T, Jiao F, Biskup E, Pan Y, Liu M, Wang L. A phase I study of the safety and activity of K-001 in patients with advanced pancreatic ductal adenocarcinoma. BMC Cancer 2021; 21:672. [PMID: 34098895 PMCID: PMC8183060 DOI: 10.1186/s12885-021-08375-6] [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: 12/06/2020] [Accepted: 05/18/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease that lack of effective therapeutic drugs. K-001 is an oral antitumor drug made from active ingredients of marine microorganisms. The current study aimed to evaluate safety and antitumor activity of K-001 in patients with advanced PDAC. METHODS In this phase I, open-label trial, patients with advanced PDAC were recruited to a dose-escalation study in a standard 3 + 3 design. K-001 was administered twice daily in four-week cycles, and dose escalation from 1350 mg to 2160 mg was evaluated twice daily. Physical examination and laboratory tests were done at screening and then weekly. The safety, dose-limiting toxicity (DLT), and maximum tolerated dose (MTD) of K-001 were assessed while tumor response was estimated by Response Evaluation Criteria in Solid Tumor (RECIST). RESULTS Eighteen patients with advanced PDAC were screened, and twelve eligible patients were analyzed in the study. No DLT was observed. Totally, 47 adverse events (AEs) presented, and 14 drug-related AEs were reported in 7 patients, including 8 grade 1 events (57.1%) and 6 grade 2 events (42.9%). There was no grade 3 or 4 drug-related AE. In these 14 drug-related AEs, the most frequent ones were dyspepsia (21.4%), followed by flatulence, constipation, and hemorrhoid bleeding (above 10% of each). Among all 12 patients, 10 patients (83.3%) maintained stable disease (SD), and 2 patients (16.7%) had progressive disease (PD). The objective response rate (ORR) was 0% and the disease control rate (DCR) was 83.3%. CONCLUSIONS K-001 manifests satisfactory safety and tolerability, as well as meaningful antitumor activity in advanced PDAC patients. Further evaluation of K-001 in phase II/III appears warranted. TRIAL REGISTRATION NCT02720666 . Registered 28 Match 2016 - Retrospectively registered.
Collapse
Affiliation(s)
- Jiujie Cui
- Department of Medical Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory of Oncogene and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Oncology, First People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Haiyan Yang
- Department of Medical Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory of Oncogene and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Oncology, First People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jue Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Donghui Chen
- Department of Oncology, First People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jiong Hu
- Department of Medical Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory of Oncogene and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Oncology, First People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Haiyan Zhang
- Department of Oncology, First People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yu Wang
- Department of Medical Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory of Oncogene and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Oncology, First People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Ting Han
- Department of Medical Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory of Oncogene and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Oncology, First People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Tiebo Mao
- Department of Medical Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory of Oncogene and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Jiao
- Department of Medical Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory of Oncogene and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Oncology, First People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Ewelina Biskup
- College of Fundamental Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Yaotian Pan
- Institute of Psychology Chinese Academy of Sciences, Beijing, China.
| | - Min Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China.
| | - Liwei Wang
- Department of Medical Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- State Key Laboratory of Oncogene and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Department of Oncology, First People's Hospital, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
19
|
Laoveeravat P, Abhyankar PR, Brenner AR, Gabr MM, Habr FG, Atsawarungruangkit A. Artificial intelligence for pancreatic cancer detection: Recent development and future direction. Artif Intell Gastroenterol 2021; 2:56-68. [DOI: 10.35712/aig.v2.i2.56] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/31/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023] Open
Abstract
Artificial intelligence (AI) has been increasingly utilized in medical applications, especially in the field of gastroenterology. AI can assist gastroenterologists in imaging-based testing and prediction of clinical diagnosis, for examples, detecting polyps during colonoscopy, identifying small bowel lesions using capsule endoscopy images, and predicting liver diseases based on clinical parameters. With its high mortality rate, pancreatic cancer can highly benefit from AI since the early detection of small lesion is difficult with conventional imaging techniques and current biomarkers. Endoscopic ultrasound (EUS) is a main diagnostic tool with high sensitivity for pancreatic adenocarcinoma and pancreatic cystic lesion. The standard tumor markers have not been effective for diagnosis. There have been recent research studies in AI application in EUS and novel biomarkers to early detect and differentiate malignant pancreatic lesions. The findings are impressive compared to the available traditional methods. Herein, we aim to explore the utility of AI in EUS and novel serum and cyst fluid biomarkers for pancreatic cancer detection.
Collapse
Affiliation(s)
- Passisd Laoveeravat
- Division of Digestive Diseases and Nutrition, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Priya R Abhyankar
- Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Aaron R Brenner
- Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Moamen M Gabr
- Division of Digestive Diseases and Nutrition, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Fadlallah G Habr
- Division of Gastroenterology, Warren Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - Amporn Atsawarungruangkit
- Division of Gastroenterology, Warren Alpert Medical School of Brown University, Providence, RI 02903, United States
| |
Collapse
|
20
|
Feng Z, Li K, Wu Y, Peng C. Transcriptomic Profiling Identifies DCBLD2 as a Diagnostic and Prognostic Biomarker in Pancreatic Ductal Adenocarcinoma. Front Mol Biosci 2021; 8:659168. [PMID: 33834039 PMCID: PMC8021715 DOI: 10.3389/fmolb.2021.659168] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Accumulating evidence shows that the elevated expression of DCBLD2 (discoidin, CUB and LCCL domain-containing protein 2) is associated with unfavorable prognosis of various cancers. However, the correlation of DCBLD2 expression value with the diagnosis and prognosis of pancreatic ductal adenocarcinoma (PDAC) has not yet been elucidated. Methods: Univariate Cox regression analysis was used to screen robust survival-related genes. Expression pattern of selected genes was investigated in PDAC tissues and normal tissues from multiple cohorts. Kaplan–Meier (K–M) survival curves, ROC curves and calibration curves were employed to assess prognostic performance. The relationship between DCBLD2 expression and immune cell infiltrates was conducted by CIBERSORT software. Biological processes and KEGG pathway enrichment analyses were adopted to clarify the potential function of DCBLD2 in PDAC. Results: Univariate analysis, K–M survival curves and calibration curves indicated that DCBLD2 was a robust prognostic factor for PDAC with cross-cohort compatibility. Upregulation of DCBLD2 was observed in dissected PDAC tissues as well as extracellular vesicles from both plasma and serum samples of PDAC patients. Both DCBLD2 expression in tissue and extracellular vesicles had significant diagnostic value. Besides, DCBLD2 expression was correlated with infiltrating level of CD8+ T cells and macrophage M2 cells. Functional enrichment revealed that DCBLD2 might be involved in cell motility, angiogenesis, and cancer-associated pathways. Conclusion: Our study systematically analyzed the potential diagnostic, prognostic and therapeutic value of DCBLD2 in PDAC. All the findings indicated that DCBLD2 might play a considerably oncogenic role in PDAC with diagnostic, prognostic and therapeutic potential. These preliminary results of bioinformatics analyses need to be further validated in more prospective studies.
Collapse
Affiliation(s)
- Zengyu Feng
- 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.,Department of General Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kexian 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
| | - Yulian Wu
- Department of General Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chenghong Peng
- 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
| |
Collapse
|