1
|
Wang X, Che Y, Liu S, Ma B, Liu Y, Lei J, Yuan L, Zhou Y, Ying J, Zhang Y, Tian C, Zhu T, Qi L, Jiang Y, Fang X. Single-Molecule Liquid Biopsy Detects Low- and High-Abundance Protein Markers Simultaneously for Pancreatic Cancer Diagnosis. Anal Chem 2025; 97:8385-8393. [PMID: 40194997 DOI: 10.1021/acs.analchem.4c07031] [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: 04/09/2025]
Abstract
Simultaneous analysis of multiple biomarkers can typically improve the sensitivity and specificity of a disease diagnosis. Low-abundance serum proteins have recently emerged as a novel class of biomarkers for diseases. Due to the low concentration, the low-abundance protein analysis relies on single-molecule immunoassay, which has a very limited dynamic range. As a result, simultaneous analysis of low- and high-abundance protein markers requires multiple instruments, which demands larger sample volumes and is cost-/labor-consuming. To overcome these limitations, we developed a single-molecule imaging technique that can detect low- and high-abundance protein markers simultaneously in one chip. By employing a hybrid biomarker capture strategy that involves both glass surface and bead immobilization, our method greatly extended the detection range of the single-molecule assay. We used the method for pancreatic cancer diagnosis and analyzed three serum biomarkers of different abundances, including LIF, CA19-9, and CA125. Combined analysis of the three biomarkers yielded exceptional sensitivity and specificity (AUC = 0.996), which is better than using any of the markers alone, including CA19-9 that is used in clinical practice (AUC = 0.804). Overall, we demonstrated a simple and cost-effective method that greatly extended the dynamic range of single-molecule imaging while maintaining the sensitivity, which has great potential in various clinical applications.
Collapse
Affiliation(s)
- Xinjian Wang
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300350, P. R. China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Yudong Che
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P. R. China
| | - Songlin Liu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Bochen Ma
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Yijun Liu
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300350, P. R. China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Jiao Lei
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Liang Yuan
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, P. R. China
| | - Yiwen Zhou
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P. R. China
| | - Jieer Ying
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P. R. China
| | - Yimin Zhang
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P. R. China
| | - Chen Tian
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, P. R. China
| | - Tao Zhu
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P. R. China
| | - Lubin Qi
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Yifei Jiang
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300350, P. R. China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, P. R. China
| | - Xiaohong Fang
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300350, P. R. China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, P. R. China
| |
Collapse
|
2
|
Lee J, Cheong H. The Role of A20 in Cancer: Friend or Foe? Cells 2025; 14:544. [PMID: 40214497 PMCID: PMC11988600 DOI: 10.3390/cells14070544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
A20 is a ubiquitin-editing enzyme that has emerged as a key regulator of inflammatory signaling with paradoxical roles in cancer. Acting as both an oncogene and a tumor suppressor gene depending on the cellular context, A20 modulates important cell pathways, such as NF-κB signaling and autophagy. In this review, we summarize the dual roles of A20 in tumorigenesis, highlighting its ability to promote tumor progression in cancers, such as breast and melanoma, while functioning as a tumor suppressor in lymphomas and hepatocellular carcinoma. We discuss the interplay of A20 with autophagy, a process that is important for maintaining cellular homeostasis and influencing tumor dynamics. By integrating recent findings, we provide insight into how dysregulation of A20 and its associated pathways can either suppress or drive cancer development, which may lead to improved therapeutic intervention.
Collapse
Affiliation(s)
| | - Heesun Cheong
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang-si 10408, Republic of Korea;
| |
Collapse
|
3
|
Koc DC, Mănescu IB, Mănescu M, Dobreanu M. A Review of the Prognostic Significance of Neutrophil-to-Lymphocyte Ratio in Nonhematologic Malignancies. Diagnostics (Basel) 2024; 14:2057. [PMID: 39335736 PMCID: PMC11431542 DOI: 10.3390/diagnostics14182057] [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: 07/01/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Biomarkers are crucial in cancer diagnostics, prognosis, and surveillance. Extensive research has been dedicated to identifying biomarkers that are broadly applicable across multiple cancer types and can be easily obtained from routine investigations such as blood cell counts. One such biomarker, the neutrophil-to-lymphocyte ratio (NLR), has been established as a prognostic marker in cancer. However, due to the dynamic nature of cancer diagnosis and treatment, periodic updates are necessary to keep abreast of the vast amount of published data. In this review, we searched the PubMed database and analyzed and synthesized recent literature (2018-February 2024) on the role of NLR in predicting clinical outcomes in nonhematologic malignancies. The search was conducted using the PubMed database. We included a total of 88 studies, encompassing 28,050 human subjects, and categorized the findings into four major groups: gastrointestinal cancer, cancers of the urinary tract and reproductive system, lung cancer, and breast cancer. Our analysis confirms that NLR is a reliable prognostic indicator in cancer, and we discuss the specific characteristics, limitations, and exceptions associated with its use. The review concludes with a concise Q&A section, presenting the most relevant take-home messages in response to five key practical questions on this topic.
Collapse
Affiliation(s)
- Defne Cigdem Koc
- Medical Campus Hamburg, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 11-15 Albert-Einstein-Ring, 22761 Hamburg, Germany; (D.C.K.); (I.B.M.)
| | - Ion Bogdan Mănescu
- Medical Campus Hamburg, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 11-15 Albert-Einstein-Ring, 22761 Hamburg, Germany; (D.C.K.); (I.B.M.)
- Department of Laboratory Medicine, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 38 Gheorghe Marinescu, 540142 Targu Mures, Romania
| | - Măriuca Mănescu
- Department of Pediatrics, Emergency County Clinical Hospital of Targu Mures, 50 Gheorghe Marinescu, 540136 Targu Mures, Romania
| | - Minodora Dobreanu
- Department of Laboratory Medicine, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 38 Gheorghe Marinescu, 540142 Targu Mures, Romania
- Clinical Laboratory, Emergency County Clinical Hospital of Targu Mures, 50 Gheorghe Marinescu, 540136 Targu Mures, Romania
| |
Collapse
|
4
|
Dixit S, Mahakalkar C, Kshirsagar S, Hatewar A. Exploring the Prognostic Role of D-dimer Levels in Pancreatic Cancer: A Comprehensive Review of Clinicopathological Associations. Cureus 2024; 16:e68627. [PMID: 39371859 PMCID: PMC11451093 DOI: 10.7759/cureus.68627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024] Open
Abstract
Pancreatic cancer is known for its dismal prognosis and high mortality rate, primarily due to late-stage diagnosis and aggressive disease progression. Finding reliable prognostic biomarkers is crucial in improving patient outcomes and guiding treatment strategies. D-dimer, a fibrin degradation product, has emerged as a potential biomarker of interest in various cancers due to its association with coagulation abnormalities. This comprehensive review investigates the prognostic role of D-dimer levels in pancreatic cancer by synthesizing current research and exploring its clinicopathological associations. Elevated D-dimer levels in pancreatic cancer patients have been linked to poorer clinical outcomes, including reduced overall survival and increased disease progression. The review examines how D-dimer levels correlate with tumor characteristics such as stage, grade, and metastatic spread, highlighting its potential utility as a prognostic marker. Additionally, the review addresses the methodological challenges in D-dimer measurement and the need for standardized protocols to enhance the reliability and applicability of results. Future research directions are identified, focusing on validating D-dimer's clinical utility and integrating it into routine practice for risk stratification and personalized treatment planning. By providing a comprehensive overview of D-dimer's prognostic value, this review aims to contribute to developing more effective management strategies for pancreatic cancer, ultimately improving patient care and outcomes.
Collapse
Affiliation(s)
- Sparsh Dixit
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Chandrashekhar Mahakalkar
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Shivani Kshirsagar
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Akansha Hatewar
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
5
|
Wu B, Zhang G, Zhao X, Wang J, Wang D, Zhang Y, Yu M, Cai H. Association between D-dimer levels and clinicopathological characteristics of pancreatic cancer and its role in prognosis: A systematic review and meta-analysis. Asian J Surg 2024; 47:3417-3424. [PMID: 38453610 DOI: 10.1016/j.asjsur.2024.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/23/2024] [Accepted: 02/02/2024] [Indexed: 03/09/2024] Open
Abstract
According to previous studies, D-dimer levels are associated with the prognosis of patients with pancreatic cancer (PC). However, the results of current studies are limited and controversial. Therefore, we performed this meta-analysis to assess the relationship between D-dimer levels and prognostic and pathological characteristics of PC patients. We first searched the databases of PubMed, Embase, The Cochrane Library, Web Of Science, CBM, VIP, CNKI and Wanfang to identify available studies. The relationship between pretreatment d-dimer levels and prognosis in PC patients was assessed using the combined hazard ratio (HR) and 95% confidence interval (CI). The combined odds ratio (OR) and 95% confidence interval (CI) were used in assessing the relationship between pathological characteristics and d-dimer levels in PC patients. Stata 12.0 software was used for all statistical analyses. In total, we included 13 studies involving 2777 patients. The results showed that elevated pre-treatment d -dimer levels were significantly associated with OS deterioration (HR = 1.46 95% CI: 1.34-1.59; p < 0.001). We also performed subgroup analyses based on sample size, d -dimer threshold, follow-up time, and HR source to further validate the prognostic value of pretreatment d -dimer levels in PC. In addition, according to the analysis, high pretreatment d -dimer levels in PC patients were associated with late tumor stage (OR = 4.78, 95% CI 1.73-13.20, p < 0. 005), larger tumor size (OR = 1.72, 95% CI 1.25-2.35, p < 0.005), and distant metastasis of tumor (OR = 5.06, 95% CI 2.45-10.43, p < 0.005) were significantly associated. In contrast, other clinicopathological factors, including age, gender and lymph node metastasis, were not associated with d-dimer levels. In conclusion, we found that high pre-treatment d-dimer levels were associated with a poor prognosis in PC patients, in relation to later tumor stage, larger tumor size and the development of distant metastases. Plasma d-dimer levels can be used as a biomarker of prognosis in PC patients.
Collapse
Affiliation(s)
- Bin Wu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China; Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China; General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Guangming Zhang
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China; General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China; The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Xiashuang Zhao
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China; Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China; General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Jie Wang
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China; Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China; General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Da Wang
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China; General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China; Jiangsu University, Zhenjiang, 212000, China
| | - Yipeng Zhang
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China; Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China; General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Miao Yu
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China; General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China; Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Hui Cai
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China; Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China; General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China; Gansu Provincial Hospital, Lanzhou, 730000, China; The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
6
|
Xia DQ, Zhou Y, Yang S, Li FF, Tian LY, Li YH, Xu HY, Xiao CZ, Wang W. Combining prognostic value of serum carbohydrate antigen 19-9 and tumor size reduction ratio in pancreatic ductal adenocarcinoma. World J Gastrointest Oncol 2024; 16:798-809. [PMID: 38577439 PMCID: PMC10989379 DOI: 10.4251/wjgo.v16.i3.798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/15/2023] [Accepted: 01/27/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a common cancer with increasing morbidity and mortality due to changes of social environment. AIM To evaluate the significance of serum carbohydrate antigen 19-9 (CA19-9) and tumor size changes pre- and post-neoadjuvant therapy (NAT). METHODS This retrospective study was conducted at the Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital. This study specifically assessed CA19-9 levels and tumor size before and after NAT. RESULTS A total of 156 patients who completed NAT and subsequently underwent tumor resection were included in this study. The average age was 65.4 ± 10.6 years and 72 (46.2%) patients were female. Before survival analysis, we defined the post-NAT serum CA19-9 level/pre-NAT serum CA19-9 level as the CA19-9 ratio (CR). The patients were divided into three groups: CR < 0.5, CR > 0.5 and < 1 and CR > 1. With regard to tumor size measured by both computed tomography and magnetic resonance imaging, we defined the post-NAT tumor size/pre-NAT tumor size as the tumor size ratio (TR). The patients were then divided into three groups: TR < 0.5, TR > 0.5 and < 1 and TR > 1. Based on these groups divided according to CR and TR, we performed both overall survival (OS) and disease-free survival (DFS) analyses. Log-rank tests showed that both OS and DFS were significantly different among the groups according to CR and TR (P < 0.05). CR and TR after NAT were associated with increased odds of achieving a complete or near-complete pathologic response. Moreover, CR (hazard ratio: 1.721, 95%CI: 1.373-3.762; P = 0.006), and TR (hazard ratio: 1.435, 95%CI: 1.275-4.363; P = 0.014) were identified as independent factors associated with OS. CONCLUSION This study demonstrated that post-NAT serum CA19-9 level/pre-NAT serum CA19-9 level and post-NAT tumor size/pre-NAT tumor size were independent factors associated with OS in patients with PDAC who received NAT and subsequent surgical resection.
Collapse
Affiliation(s)
- Dong-Qin Xia
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yong Zhou
- Department of Oncology, Chongqing Weisiteng Biotech Translational Research Institute, Chongqing 430039, China
- Chongqing Key Laboratory on Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 430065, China
| | - Shuang Yang
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Fang-Fei Li
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Li-Ya Tian
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yan-Hua Li
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Hai-Yan Xu
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Cai-Zhi Xiao
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Wei Wang
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
7
|
Chowdhury NN, Yang Y, Dutta A, Luo M, Wei Z, Abrahams SR, Revenko AS, Shah F, Miles LA, Parmer RJ, de Laat B, Wolberg AS, Luyendyk JP, Fishel ML, Flick MJ. Plasminogen deficiency suppresses pancreatic ductal adenocarcinoma disease progression. Mol Oncol 2024; 18:113-135. [PMID: 37971174 PMCID: PMC10766200 DOI: 10.1002/1878-0261.13552] [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: 12/13/2022] [Revised: 10/06/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly fatal metastatic disease associated with robust activation of the coagulation and fibrinolytic systems. However, the potential contribution of the primary fibrinolytic protease plasminogen to PDAC disease progression has remained largely undefined. Mice bearing C57Bl/6-derived KPC (KRasG12D , TRP53R172H ) tumors displayed evidence of plasmin activity in the form of high plasmin-antiplasmin complexes and high plasmin generation potential relative to mice without tumors. Notably, plasminogen-deficient mice (Plg- ) had significantly diminished KPC tumor growth in subcutaneous and orthotopic implantation models. Moreover, the metastatic potential of KPC cells was significantly diminished in Plg- mice, which was linked to reduced early adhesion and/or survival of KPC tumor cells. The reduction in primary orthotopic KPC tumor growth in Plg- mice was associated with increased apoptosis, reduced accumulation of pro-tumor immune cells, and increased local proinflammatory cytokine production. Elimination of fibrin(ogen), the primary proteolytic target of plasmin, did not alter KPC primary tumor growth and resulted in only a modest reduction in metastatic potential. In contrast, deficiencies in the plasminogen receptors Plg-RKT or S100A10 in tumor cells significantly reduced tumor growth. Plg-RKT reduction in tumor cells, but not reduced S100A10, suppressed metastatic potential in a manner that mimicked plasminogen deficiency. Finally, tumor growth was also reduced in NSG mice subcutaneously or orthotopically implanted with patient-derived PDAC tumor cells in which circulating plasminogen was pharmacologically reduced. Collectively, these studies suggest that plasminogen promotes PDAC tumor growth and metastatic potential, in part through engaging plasminogen receptors on tumor cells.
Collapse
Affiliation(s)
- Nayela N. Chowdhury
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndianapolisINUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisINUSA
| | - Yi Yang
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillNCUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillNCUSA
- UNC Blood Research CenterUniversity of North Carolina at Chapel HillNCUSA
| | - Ananya Dutta
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillNCUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillNCUSA
- UNC Blood Research CenterUniversity of North Carolina at Chapel HillNCUSA
| | - Michelle Luo
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillNCUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillNCUSA
- UNC Blood Research CenterUniversity of North Carolina at Chapel HillNCUSA
| | - Zimu Wei
- Department of Pathobiology & Diagnostic InvestigationMichigan State UniversityEast LansingMIUSA
- Institute for Integrative ToxicologyMichigan State UniversityEast LansingMIUSA
- Department of Pharmacology and ToxicologyMichigan State UniversityEast LansingMIUSA
| | - Sara R. Abrahams
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillNCUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillNCUSA
- UNC Blood Research CenterUniversity of North Carolina at Chapel HillNCUSA
| | | | - Fenil Shah
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndianapolisINUSA
| | - Lindsey A. Miles
- Department of Molecular MedicineScripps Research InstituteLa JollaCAUSA
| | - Robert J. Parmer
- Department of Medicine, Veterans Administration San Diego Healthcare SystemUniversity of California, San DiegoCAUSA
| | - Bas de Laat
- Synapse Research InstituteMaastrichtThe Netherlands
| | - Alisa S. Wolberg
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillNCUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillNCUSA
- UNC Blood Research CenterUniversity of North Carolina at Chapel HillNCUSA
| | - James P. Luyendyk
- Department of Pathobiology & Diagnostic InvestigationMichigan State UniversityEast LansingMIUSA
- Institute for Integrative ToxicologyMichigan State UniversityEast LansingMIUSA
- Department of Pharmacology and ToxicologyMichigan State UniversityEast LansingMIUSA
| | - Melissa L. Fishel
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndianapolisINUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisINUSA
| | - Matthew J. Flick
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillNCUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillNCUSA
- UNC Blood Research CenterUniversity of North Carolina at Chapel HillNCUSA
| |
Collapse
|
8
|
González-Borja I, Viúdez A, Alors-Pérez E, Goñi S, Amat I, Ghanem I, Pazo-Cid R, Feliu J, Alonso L, López C, Arrazubi V, Gallego J, Pérez-Sanz J, Hernández-García I, Vera R, Castaño JP, Fernández-Irigoyen J. Cytokines and Lymphoid Populations as Potential Biomarkers in Locally and Borderline Pancreatic Adenocarcinoma. Cancers (Basel) 2022; 14:5993. [PMID: 36497475 PMCID: PMC9739487 DOI: 10.3390/cancers14235993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/09/2022] Open
Abstract
Despite its relative low incidence, PDAC is one of the most aggressive and lethal types of cancer, being currently the seventh leading cause of cancer death worldwide, with a 5-year survival rate of 10.8%. Taking into consideration the necessity to improve the prognosis of these patients, this research has been focused on the discovery of new biomarkers. For this purpose, patients with BL and resectable disease were recruited. Serum cytokines and growth factors were monitored at different time points using protein arrays. Immune cell populations were determined by flow cytometry in peripheral blood as well as by immunohistochemistry (IHC) in tumor tissues. Several cytokines were found to be differentially expressed between the study subgroups. In the BL disease setting, two different scores were proven to be independent prognostic factors for progression-free survival (PFS) (based on IL-10, MDC, MIF, and eotaxin-3) and OS (based on eotaxin-3, NT-3, FGF-9, and IP10). In the same context, CA19-9 was found to play a role as independent prognostic factor for OS. Eotaxin-3 and MDC cytokines for PFS, and eotaxin-3, NT-3, and CKβ8-1 for OS, were shown to be predictive biomarkers for nab-paclitaxel and gemcitabine regimen. Similarly, oncostatin, BDNF, and IP10 cytokines were proven to act as predictive biomarkers regarding PFS, for FOLFIRINOX regimen. In the resectable cohort, RANTES, TIMP-1, FGF-4, and IL-10 individually differentiated patients according to their cancer-associated survival. Regarding immune cell populations, baseline high levels of circulating B lymphocytes were related to a significantly longer OS, while these levels significantly decreased as progression occurred. Similarly, baseline high levels of helper lymphocytes (CD4+), low levels of cytotoxic lymphocytes (CD8+), and a high CD4/CD8 ratio, were related to a significantly longer PFS. Finally, high levels of CD4+ and CD8+ intratumoural infiltration was associated with significantly longer PFS. In conclusion, in this study we were able to identify several prognostic and predictive biomarker candidates in patients diagnosed of resectable or BL PDAC.
Collapse
Affiliation(s)
- Iranzu González-Borja
- OncobionaTras Lab, Navarrabiomed, Navarra University Hospital, Universidad Pública de Navarra (UPNA), 31006 Pamplona, Spain
| | - Antonio Viúdez
- OncobionaTras Lab, Navarrabiomed, Navarra University Hospital, Universidad Pública de Navarra (UPNA), 31006 Pamplona, Spain
- Medical Oncology Department, Navarra University Hospital, 31008 Pamplona, Spain
| | - Emilia Alors-Pérez
- Maimonides Biomedical Research Institute of Córdoba, 14004 Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, 14071 Córdoba, Spain
- Reina Sofía University Hospital, 14004 Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), 14004 Córdoba, Spain
| | - Saioa Goñi
- OncobionaTras Lab, Navarrabiomed, Navarra University Hospital, Universidad Pública de Navarra (UPNA), 31006 Pamplona, Spain
| | - Irene Amat
- Pathology Department, Navarra University Hospital, 31008 Pamplona, Spain
| | - Ismael Ghanem
- Medical Oncology Department, La Paz University Hospital, 28046 Madrid, Spain
| | - Roberto Pazo-Cid
- Medical Oncology Department, Miguel Servet University Hospital, 50009 Zaragoza, Spain
| | - Jaime Feliu
- Medical Oncology Department, La Paz University Hospital, 28046 Madrid, Spain
| | - Laura Alonso
- Pathology Department, Navarra University Hospital, 31008 Pamplona, Spain
| | - Carlos López
- Medical Oncology Department, Marqués de Valdecilla University Hospital, 39008 Santander, Spain
| | - Virginia Arrazubi
- Medical Oncology Department, Navarra University Hospital, 31008 Pamplona, Spain
| | - Javier Gallego
- Medical Oncology Department, Hospital General Universitario de Elche, 03203 Elche, Spain
| | - Jairo Pérez-Sanz
- OncobionaTras Lab, Navarrabiomed, Navarra University Hospital, Universidad Pública de Navarra (UPNA), 31006 Pamplona, Spain
| | | | - Ruth Vera
- Medical Oncology Department, Navarra University Hospital, 31008 Pamplona, Spain
| | - Justo P Castaño
- Maimonides Biomedical Research Institute of Córdoba, 14004 Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, 14071 Córdoba, Spain
- Reina Sofía University Hospital, 14004 Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), 14004 Córdoba, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Navarrabiomed, Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Navarra University Hospital, Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| |
Collapse
|
9
|
Topkan E, Selek U, Pehlivan B, Kucuk A, Haksoyler V, Kilic Durankus N, Sezen D, Bolukbasi Y. The Prognostic Significance of Novel Pancreas Cancer Prognostic Index in Unresectable Locally Advanced Pancreas Cancers Treated with Definitive Concurrent Chemoradiotherapy. J Inflamm Res 2021; 14:4433-4444. [PMID: 34511977 PMCID: PMC8427684 DOI: 10.2147/jir.s329611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/18/2021] [Indexed: 12/22/2022] Open
Abstract
PURPOSE We evaluated the prognostic quality of the novel pancreas cancer prognostic index (PCPI), a combination of CA 19-9 and systemic inflammation response index (SIRI), on the outcomes of locally advanced pancreas adenocarcinoma (LAPAC) patients who received concurrent chemoradiotherapy (C-CRT). METHODS This retrospective analysis covered 152 unresectable LAPAC patients treated from 2007 to 2019. Receiver operating characteristic (ROC) curve analysis was used to define ideal cutoff thresholds for the pretreatment CA 19-9 and SIRI measurements, individually. The associations between the PCPI groups and progression-free- (PFS) and overall survival (OS) comprised the respective primary and secondary endpoints. RESULTS The ROC curve analysis distinguished the respective rounded optimal cutoffs at 91 U/m/L (< versus ≥90) and 1.8 (< versus ≥1.8) for CA 19-9 and SIRI, arranging the study cohort into two significantly different survival groups for each, with resultant four likely groups: Group-1: CA 19-9<90 U/m/L and SIRI<1.8, Group-2: CA 19-9<90 U/m/L but SIRI≥1.8, Group-3: CA 19-9≥90 U/m/L but SIRI<1.8, and Group-4: CA 19-9≥90 U/m/L and SIRI≥1.8. Since the PFS (P=0.79) and OS (P=0.86) estimates of the groups 2 and 3 were statistically indistinct, we merged them as one group and created the novel three-tiered PCPI: PCPI-1: CA 19-9<90 U/m/L and SIRI<1.8, PCPI-2: CA 19-9<90 U/m/L but SIRI≥1.8 or CA 19-9≥90 U/m/L but SIRI<1.8, and PCPI-3: CA 19-9≥90 U/m/L and SIRI≥1.8, respectively. Comparative analyses unveiled that the PCPI-1 and PCPI-3 groups had the respective best and worst PFS (17.0 versus 7.5 versus 4.4 months; P<0.001) and OS (26.1 versus 15.1 versus 7.4 months; P<0.001) outcomes, while the PCPI-2 group posed in between. The multivariate analysis outcomes confirmed the novel three tired PCPI's independent prognostic significance on either of the PFS [HR: 5.38 (95% confidence interval (CI): 4.96-5.80); P<0.001)] and OS [HR: 5.67 (95% CI: 5.19-6.15); P<0.001] endpoints, separately. CONCLUSION The new PCPI introduced here can be used as an independent and reliable prognostic indicator to divide LAPAC patients into three subgroups with discrete survival results.
Collapse
Affiliation(s)
- Erkan Topkan
- Department of Radiation Oncology, Baskent University Medical Faculty, Adana, Turkey
| | - Ugur Selek
- Department of Radiation Oncology, Koc University Faculty of Medicine, Istanbul, Turkey
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Berrin Pehlivan
- Department of Radiation Oncology, Bahcesehir University, Istanbul, Turkey
| | - Ahmet Kucuk
- Radiation Oncology Clinics, Mersin City Hospital, Mersin, Turkey
| | | | | | - Duygu Sezen
- Department of Radiation Oncology, Koc University Faculty of Medicine, Istanbul, Turkey
| | - Yasemin Bolukbasi
- Department of Radiation Oncology, Koc University Faculty of Medicine, Istanbul, Turkey
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|