1
|
Kim H, Huh S, Park J, Han Y, Ahn KG, Noh Y, Lee SJ, Chu H, Kim SS, Jung HS, Yun WG, Cho YJ, Kwon W, Jang JY, Kang UB. Development of a Fit-For-Purpose Multi-Marker Panel for Early Diagnosis of Pancreatic Ductal Adenocarcinoma. Mol Cell Proteomics 2024; 23:100824. [PMID: 39097268 PMCID: PMC11406441 DOI: 10.1016/j.mcpro.2024.100824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/28/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) suffers from a lack of an effective diagnostic method, which hampers improvement in patient survival. Carbohydrate antigen 19-9 (CA19-9) is the only FDA-approved blood biomarker for PDAC, yet its clinical utility is limited due to suboptimal performance. Liquid chromatography-mass spectrometry (LC-MS) has emerged as a burgeoning technology in clinical proteomics for the discovery, verification, and validation of novel biomarkers. A plethora of protein biomarker candidates for PDAC have been identified using LC-MS, yet few has successfully transitioned into clinical practice. This translational standstill is owed partly to insufficient considerations of practical needs and perspectives of clinical implementation during biomarker development pipelines, such as demonstrating the analytical robustness of proposed biomarkers which is critical for transitioning from research-grade to clinical-grade assays. Moreover, the throughput and cost-effectiveness of proposed assays ought to be considered concomitantly from the early phases of the biomarker pipelines for enhancing widespread adoption in clinical settings. Here, we developed a fit-for-purpose multi-marker panel for PDAC diagnosis by consolidating analytically robust biomarkers as well as employing a relatively simple LC-MS protocol. In the discovery phase, we comprehensively surveyed putative PDAC biomarkers from both in-house data and prior studies. In the verification phase, we developed a multiple-reaction monitoring (MRM)-MS-based proteomic assay using surrogate peptides that passed stringent analytical validation tests. We adopted a high-throughput protocol including a short gradient (<10 min) and simple sample preparation (no depletion or enrichment steps). Additionally, we developed our assay using serum samples, which are usually the preferred biospecimen in clinical settings. We developed predictive models based on our final panel of 12 protein biomarkers combined with CA19-9, which showed improved diagnostic performance compared to using CA19-9 alone in discriminating PDAC from non-PDAC controls including healthy individuals and patients with benign pancreatic diseases. A large-scale clinical validation is underway to demonstrate the clinical validity of our novel panel.
Collapse
Affiliation(s)
- Hyeonji Kim
- Bertis R&D Division, Bertis Inc, Gyeonggi-do, Republic of Korea
| | - Sunghyun Huh
- Bertis R&D Division, Bertis Inc, Gyeonggi-do, Republic of Korea
| | | | - Youngmin Han
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyung-Geun Ahn
- Bertis R&D Division, Bertis Inc, Gyeonggi-do, Republic of Korea
| | - Yiyoung Noh
- Bertis R&D Division, Bertis Inc, Gyeonggi-do, Republic of Korea
| | - Seong-Jae Lee
- Bertis R&D Division, Bertis Inc, Gyeonggi-do, Republic of Korea
| | - Hyosub Chu
- Bertis R&D Division, Bertis Inc, Gyeonggi-do, Republic of Korea
| | - Sung-Soo Kim
- Manufacturing and Technology Division, Bertis Inc, Gyeonggi-do, Republic of Korea
| | - Hye-Sol Jung
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Won-Gun Yun
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young Jae Cho
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Un-Beom Kang
- Bertis R&D Division, Bertis Inc, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
2
|
Przybyszewski O, Mik M, Nowicki M, Kusiński M, Mikołajczyk-Solińska M, Śliwińska A. Using microRNAs Networks to Understand Pancreatic Cancer-A Literature Review. Biomedicines 2024; 12:1713. [PMID: 39200178 PMCID: PMC11351910 DOI: 10.3390/biomedicines12081713] [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: 05/30/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 09/02/2024] Open
Abstract
Pancreatic cancer is a severe disease, challenging to diagnose and treat, and thereby characterized by a poor prognosis and a high mortality rate. Pancreatic ductal adenocarcinoma (PDAC) represents approximately 90% of pancreatic cancer cases, while other cases include neuroendocrine carcinoma. Despite the growing knowledge of the pathophysiology of this cancer, the mortality rate caused by it has not been effectively reduced. Recently, microRNAs have aroused great interest among scientists and clinicians, as they are negative regulators of gene expression, which participate in many processes, including those related to the development of pancreatic cancer. The aim of this review is to show how microRNAs (miRNAs) affect key signaling pathways and related cellular processes in pancreatic cancer development, progression, diagnosis and treatment. We included the results of in vitro studies, animal model of pancreatic cancer and those performed on blood, saliva and tumor tissue isolated from patients suffering from PDAC. Our investigation identified numerous dysregulated miRNAs involved in KRAS, JAK/STAT, PI3/AKT, Wnt/β-catenin and TGF-β signaling pathways participating in cell cycle control, proliferation, differentiation, apoptosis and metastasis. Moreover, some miRNAs (miRNA-23a, miRNA-24, miRNA-29c, miRNA-216a) seem to be engaged in a crosstalk between signaling pathways. Evidence concerning the utility of microRNAs in the diagnosis and therapy of this cancer is poor. Therefore, despite growing knowledge of the involvement of miRNAs in several processes associated with pancreatic cancer, we are beginning to recognize and understand their role and usefulness in clinical practice.
Collapse
Affiliation(s)
- Oskar Przybyszewski
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska St., 92-213 Lodz, Poland
| | - Michał Mik
- Department of General and Colorectal Surgery, Medical University of Lodz, 113 Stefana Żeromskiego St., 90-549 Lodz, Poland; (M.M.); (M.N.)
| | - Michał Nowicki
- Department of General and Colorectal Surgery, Medical University of Lodz, 113 Stefana Żeromskiego St., 90-549 Lodz, Poland; (M.M.); (M.N.)
| | - Michał Kusiński
- Department of Endocrinological, General and Oncological Surgery, Medical University of Lodz, 62 Pabianicka St., 93-513 Lodz, Poland;
| | - Melania Mikołajczyk-Solińska
- Department of Internal Medicine, Diabetology and Clinical Pharmacology, Medical University of Lodz, 251 Pomorska St., 92-213 Lodz, Poland;
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska St., 92-213 Lodz, Poland
| |
Collapse
|
3
|
Dias E Silva D, Chung V. Neoadjuvant treatment for pancreatic cancer: Controversies and advances. Cancer Treat Res Commun 2024; 39:100804. [PMID: 38508132 DOI: 10.1016/j.ctarc.2024.100804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/28/2024] [Accepted: 03/02/2024] [Indexed: 03/22/2024]
Abstract
Despite the advancements in the treatment of localized pancreatic cancer, several unresolved issues persist in clinical practice, especially in the neoadjuvant setting. These include determining the criteria for selecting patients for treatment, identifying the most effective chemotherapy regimens, understanding the role of radiotherapy, and accurately assessing how patients respond to treatment. Current strategies for assessing patients before surgery involve thoroughly evaluating their overall health status, analyzing tumor markers, and using advanced imaging techniques. However, existing methods for staging the disease still have limitations when it comes to accurately detecting metastatic cancer. The ongoing debate between performing surgery upfront or administering neoadjuvant therapy highlights the need for robust clinical evidence to guide treatment decisions effectively. This review analyzes the evidence regarding controversial topics in neoadjuvant pancreatic cancer treatment and discusses further research efforts to enhance patient outcomes. To improve the outcomes found with surgery alone, multimodal treatment with chemotherapy.
Collapse
Affiliation(s)
| | - Vincent Chung
- City of Hope, 1500 E. Duarte Road, Duarte, CA 91010, United States.
| |
Collapse
|
4
|
Young Han C, Bedia JS, Yang WL, Hawley SJ, Bergan L, Hopper M, Celestino J, Guo J, Gornet TG, Soosaipillai A, Yang H, Doskocil SD, Lokshin AE, Handy BC, Diamandis EP, Moore RG, Lu KH, Lu Z, Anderson KS, Drescher CW, Skates SJ, Bast RC. Autoantibodies, antigen-autoantibody complexes and antigens complement CA125 for early detection of ovarian cancer. Br J Cancer 2024; 130:861-868. [PMID: 38195887 PMCID: PMC10912308 DOI: 10.1038/s41416-023-02560-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: 03/04/2023] [Revised: 11/23/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Multiple antigens, autoantibodies (AAb), and antigen-autoantibody (Ag-AAb) complexes were compared for their ability to complement CA125 for early detection of ovarian cancer. METHODS Twenty six biomarkers were measured in a single panel of sera from women with early stage (I-II) ovarian cancers (n = 64), late stage (III-IV) ovarian cancers (186), benign pelvic masses (200) and from healthy controls (502), and then split randomly (50:50) into a training set to identify the most promising classifier and a validation set to compare its performance to CA125 alone. RESULTS Eight biomarkers detected ≥ 8% of early stage cases at 98% specificity. A four-biomarker panel including CA125, HE4, HE4 Ag-AAb and osteopontin detected 75% of early stage cancers in the validation set from among healthy controls compared to 62% with CA125 alone (p = 0.003) at 98% specificity. The same panel increased sensitivity for distinguishing early-stage ovarian cancers from benign pelvic masses by 25% (p = 0.0004) at 95% specificity. From 21 autoantibody candidates, 3 AAb (anti-p53, anti-CTAG1 and annt-Il-8) detected 22% of early stage ovarian cancers, potentially lengthening lead time prior to diagnosis. CONCLUSION A four biomarker panel achieved greater sensitivity at the same specificity for early detection of ovarian cancer than CA125 alone.
Collapse
Affiliation(s)
- Chae Young Han
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jacob S Bedia
- Biostatistics Center, Massachusetts General Hospital, Boston, MA, USA
| | - Wei-Lei Yang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarah J Hawley
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lindsay Bergan
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Marika Hopper
- Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Joseph Celestino
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Guo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Terrie G Gornet
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Hailing Yang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samantha D Doskocil
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anna E Lokshin
- Departments of Pathology, Medicine, and Obstetrics and Gynecology, University of Pittsburgh Medical Center and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Beverly C Handy
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Richard G Moore
- Department of Obstetrics and Gynecology, Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Karen H Lu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhen Lu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Charles W Drescher
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| | - Steven J Skates
- Biostatistics Center, Massachusetts General Hospital, Boston, MA, USA.
| | - Robert C Bast
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
5
|
Guo M, Zhao H. Growth differentiation factor-15 may be a novel biomarker in pancreatic cancer: A review. Medicine (Baltimore) 2024; 103:e36594. [PMID: 38335385 PMCID: PMC10860926 DOI: 10.1097/md.0000000000036594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 02/12/2024] Open
Abstract
Pancreatic cancer is a highly malignant and invasive gastrointestinal tumor that is often diagnosed at an advanced stage with a poor prognosis and high mortality. Currently, carbohydrate antigen199(CA199) is the only biomarker approved by the FDA for the diagnosis of pancreatic cancer, but it has great limitations. Growth differentiation factor-15 (GDF-15) is expected to be a novel biomarker for the diagnosis, efficacy prediction, and prognosis assessment of pancreatic cancer patients. In this paper, we searched the keywords GDF-15, macrophage inhibitory cytokine-1 (MIC-1), CA199, pancreatic cancer, and tumor markers in PubMed and Web of Science, searched related articles, and read and analyzed the retrieved papers. Finally, we systematically described the characteristics, mechanism of action, and clinical value of GDF-15, aiming to provide help for the detection and treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Meng Guo
- Shanghai Jiaotong University School of Medicine affiliated Tongren Hospital, Shanghai, China
| | - Hui Zhao
- Shanghai Jiaotong University School of Medicine affiliated Tongren Hospital, Shanghai, China
| |
Collapse
|
6
|
Ram KSR, R A. Ensembling Model Approach for Prediction of Pancreatic Cancer Using a Biomarker Panel and Multi-Model Classifiers. 2023 INTERNATIONAL CONFERENCE ON NEXT GENERATION ELECTRONICS (NELEX) 2023:1-6. [DOI: 10.1109/nelex59773.2023.10421733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- K Saaketh Raja Ram
- School Of Computing Amrita Vishwa Vidyapeetham,Department Of CSE - AIE,Chennai,India
| | - Annamalai R
- School Of Computing, Amrita Vishwa Vidyapeetham,Department Of CSE,Chennai,India
| |
Collapse
|
7
|
Khojasteh-Leylakoohi F, Mohit R, Khalili-Tanha N, Asadnia A, Naderi H, Pourali G, Yousefli Z, Khalili-Tanha G, Khazaei M, Maftooh M, Nassiri M, Hassanian SM, Ghayour-Mobarhan M, Ferns GA, Shahidsales S, Lam AKY, Giovannetti E, Nazari E, Batra J, Avan A. Down regulation of Cathepsin W is associated with poor prognosis in pancreatic cancer. Sci Rep 2023; 13:16678. [PMID: 37794108 PMCID: PMC10551021 DOI: 10.1038/s41598-023-42928-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 09/16/2023] [Indexed: 10/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with a very poor prognosis. Therefore, there has been a focus on identifying new biomarkers for its early diagnosis and the prediction of patient survival. Genome-wide RNA and microRNA sequencing, bioinformatics and Machine Learning approaches to identify differentially expressed genes (DEGs), followed by validation in an additional cohort of PDAC patients has been undertaken. To identify DEGs, genome RNA sequencing and clinical data from pancreatic cancer patients were extracted from The Cancer Genome Atlas Database (TCGA). We used Kaplan-Meier analysis of survival curves was used to assess prognostic biomarkers. Ensemble learning, Random Forest (RF), Max Voting, Adaboost, Gradient boosting machines (GBM), and Extreme Gradient Boosting (XGB) techniques were used, and Gradient boosting machines (GBM) were selected with 100% accuracy for analysis. Moreover, protein-protein interaction (PPI), molecular pathways, concomitant expression of DEGs, and correlations between DEGs and clinical data were analyzed. We have evaluated candidate genes, miRNAs, and a combination of these obtained from machine learning algorithms and survival analysis. The results of Machine learning identified 23 genes with negative regulation, five genes with positive regulation, seven microRNAs with negative regulation, and 20 microRNAs with positive regulation in PDAC. Key genes BMF, FRMD4A, ADAP2, PPP1R17, and CACNG3 had the highest coefficient in the advanced stages of the disease. In addition, the survival analysis showed decreased expression of hsa.miR.642a, hsa.mir.363, CD22, BTNL9, and CTSW and overexpression of hsa.miR.153.1, hsa.miR.539, hsa.miR.412 reduced survival rate. CTSW was identified as a novel genetic marker and this was validated using RT-PCR. Machine learning algorithms may be used to Identify key dysregulated genes/miRNAs involved in the disease pathogenesis can be used to detect patients in earlier stages. Our data also demonstrated the prognostic and diagnostic value of CTSW in PDAC.
Collapse
Affiliation(s)
- Fatemeh Khojasteh-Leylakoohi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Mohit
- Department of Anesthesia, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Nima Khalili-Tanha
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Asadnia
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Naderi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghazaleh Pourali
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Yousefli
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghazaleh Khalili-Tanha
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Maftooh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Nassiri
- Recombinant Proteins Research Group, The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton and Sussex Medical School, Division of Medical Education, Falmer, Brighton, BN1 9PH, Sussex, UK
| | | | - Alfred King-Yin Lam
- Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast Campus, Gold Coast, QLD, 4222, Australia
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam U.M.C., VU. University Medical Center (VUMC), Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start up Unit, Fondazione Pisana Per La Scienza, Pisa, Italy
| | - Elham Nazari
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Health Information, Technology and Management, School of Allied Medical Sciences, Shahid BeheshtiUniversity of Medical Science, Tehran, Iran.
| | - Jyotsna Batra
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, 4000, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, 4102, Australia
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq.
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, 4000, Australia.
| |
Collapse
|
8
|
Saadh MJ, Baher H, Li Y, Chaitanya M, Arias-Gonzáles JL, Allela OQB, Mahdi MH, Carlos Cotrina-Aliaga J, Lakshmaiya N, Ahjel S, Amin AH, Gilmer Rosales Rojas G, Ameen F, Ahsan M, Akhavan-Sigari R. The bioengineered and multifunctional nanoparticles in pancreatic cancer therapy: Bioresponisive nanostructures, phototherapy and targeted drug delivery. ENVIRONMENTAL RESEARCH 2023; 233:116490. [PMID: 37354932 DOI: 10.1016/j.envres.2023.116490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 06/26/2023]
Abstract
The multidisciplinary approaches in treatment of cancer appear to be essential in term of bringing benefits of several disciplines and their coordination in tumor elimination. Because of the biological and malignant features of cancer cells, they have ability of developing resistance to conventional therapies such as chemo- and radio-therapy. Pancreatic cancer (PC) is a malignant disease of gastrointestinal tract in which chemotherapy and radiotherapy are main tools in its treatment, and recently, nanocarriers have been emerged as promising structures in its therapy. The bioresponsive nanocarriers are able to respond to pH and redox, among others, in targeted delivery of cargo for specific treatment of PC. The loading drugs on the nanoparticles that can be synthetic or natural compounds, can help in more reduction in progression of PC through enhancing their intracellular accumulation in cancer cells. The encapsulation of genes in the nanoparticles can protect against degradation and promotes intracellular accumulation in tumor suppression. A new kind of therapy for cancer is phototherapy in which nanoparticles can stimulate both photothermal therapy and photodynamic therapy through hyperthermia and ROS overgeneration to trigger cell death in PC. Therefore, synergistic therapy of phototherapy with chemotherapy is performed in accelerating tumor suppression. One of the important functions of nanotechnology is selective targeting of PC cells in reducing side effects on normal cells. The nanostructures are capable of being surface functionalized with aptamers, proteins and antibodies to specifically target PC cells in suppressing their progression. Therefore, a specific therapy for PC is provided and future implications for diagnosis of PC is suggested.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan; Applied Science Research Center. Applied Science Private University, Amman, Jordan
| | - Hala Baher
- Department of Radiology and Ultrasonography Techniques, College of Medical Techniques, Al-Farahidi University, Baghdad, Iraq
| | - Yuanji Li
- Institute of Electrical Engineering, Yanshan University, Qinhuangdao, 066004, China
| | - Mvnl Chaitanya
- Department of Pharmacognosy, School of Pharmacy, Lovely Professional University, Phagwara, Punjab, 144001, India
| | - José Luis Arias-Gonzáles
- Department of Social Sciences, Faculty of Social Studies, University of British Columbia, Vancouver, Canada
| | | | | | | | - Natrayan Lakshmaiya
- Department of Mechanical Engineering, Saveetha School of Engineering, SIMATS, Chennai, Tamil Nadu, India
| | - Salam Ahjel
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Ali H Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | | | - Fuad Ameen
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Muhammad Ahsan
- Department of Measurememts and Control Systems, Silesian University of Technology, Gliwice, 44-100, Poland.
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
9
|
Liang J, Tong WG. Ultrasensitive Detection and Separation of Pancreatic Cancer Biomarker CA 19-9 Using a Multiphoton Laser Wave-Mixing Detector Interfaced to Capillary Electrophoresis. ACS OMEGA 2023; 8:31030-31039. [PMID: 37663511 PMCID: PMC10468764 DOI: 10.1021/acsomega.3c02845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/19/2023] [Indexed: 09/05/2023]
Abstract
The carbohydrate antigen 19-9 (CA 19-9) is the most commonly used biomarker in the clinical diagnosis of pancreatic cancer. Multiphoton nonlinear laser wave-mixing spectroscopy is presented as an ultrasensitive detection method for CA 19-9. Wave mixing is an optical absorption-based method, and hence, one can detect CA 19-9 without labels in their native form using compact ultraviolet (UV) lasers or labeled samples using a visible laser. The wave-mixing signal exhibits a quadratic dependence on the sample concentration, and hence, it is an ideal sensor to monitor small changes in the sample. Wave mixing has inherent advantages over other absorption-based detection methods, including short optical path length (micrometer-thin samples instead of 1 cm cuvette) and excellent spatial resolution (micrometer probe). Since the laser wave-mixing probe volume is small (picoliter), it is convenient to interface to microfluidics or capillary-based electrophoresis systems to enhance chemical specificity. Our wave-mixing detectors could be configured as portable battery-powered devices suitable for field use. Laser wave-mixing spectroscopy offers enhanced selectivity levels for protein detection when coupled with capillary electrophoresis (CE). We report a concentration detection limit of 0.16 U/mL, and a corresponding mass detection limit of 1.2 × 10-8 U, and these detection limits are better than those of chemiluminescence- or ELISA- based methods.
Collapse
Affiliation(s)
- Jie Liang
- Department of Chemistry and
Biochemistry, San Diego State University, San Diego 92182, California, United
States
| | - William G. Tong
- Department of Chemistry and
Biochemistry, San Diego State University, San Diego 92182, California, United
States
| |
Collapse
|
10
|
Marin AM, Batista M, Korte de Azevedo AL, Bombardelli Gomig TH, Soares Caldeira Brant R, Chammas R, Uno M, Dias Araújo D, Zanette DL, Nóbrega Aoki M. Screening of Exosome-Derived Proteins and Their Potential as Biomarkers in Diagnostic and Prognostic for Pancreatic Cancer. Int J Mol Sci 2023; 24:12604. [PMID: 37628784 PMCID: PMC10454563 DOI: 10.3390/ijms241612604] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/11/2023] [Accepted: 07/18/2023] [Indexed: 08/27/2023] Open
Abstract
In the oncological area, pancreatic cancer is one of the most lethal diseases, with 5-year survival rising just 10% in high-development countries. This disease is genetically characterized by KRAS as a driven mutation followed by SMAD4, CDKN2, and TP53-associated mutations. In clinical aspects, pancreatic cancer presents unspecific clinical symptoms with the absence of screening and early plasmatic biomarker, being that CA19-9 is the unique plasmatic biomarker having specificity and sensitivity limitations. We analyzed the plasmatic exosome proteomic profile of 23 patients with pancreatic cancer and 10 healthy controls by using Nanoscale liquid chromatography coupled to tandem mass spectrometry (NanoLC-MS/MS). The pancreatic cancer patients were subdivided into IPMN and PDAC. Our findings show 33, 34, and 7 differentially expressed proteins when comparing the IPMN vs. control, PDAC-No treatment vs. control, and PDAC-No treatment vs. IPMN groups, highlighting proteins of the complement system and coagulation, such as C3, APOB, and SERPINA. Additionally, PDAC with no treatment showed 11 differentially expressed proteins when compared to Folfirinox neoadjuvant therapy or Gemcitabine adjuvant therapy. So here, we found plasmatic exosome-derived differentially expressed proteins among cancer patients (IPMN, PDAC) when comparing with healthy controls, which could represent alternative biomarkers for diagnostic and prognostic evaluation, supporting further scientific and clinical studies on pancreatic cancer.
Collapse
Affiliation(s)
- Anelis Maria Marin
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81350-010, Brazil; (A.M.M.); (M.B.); (D.L.Z.)
| | - Michel Batista
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81350-010, Brazil; (A.M.M.); (M.B.); (D.L.Z.)
- Mass Spectrometry Facility RPT02H, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81350-010, Brazil
| | - Alexandre Luiz Korte de Azevedo
- Laboratory of Human Cytogenetics and Oncogenetics, Genetic Department, University of Parana State (UFPR), Curitiba 80060-000, Brazil; (A.L.K.d.A.); (T.H.B.G.)
| | - Talita Helen Bombardelli Gomig
- Laboratory of Human Cytogenetics and Oncogenetics, Genetic Department, University of Parana State (UFPR), Curitiba 80060-000, Brazil; (A.L.K.d.A.); (T.H.B.G.)
| | - Rodrigo Soares Caldeira Brant
- Mass Spectrometry Facility RPT02H, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81350-010, Brazil
| | - Roger Chammas
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), Comprehensive Center for Precision Oncology (C2PO), Universidade de São Paulo, São Paulo 05508-220, Brazil; (R.C.); (M.U.); (D.D.A.)
| | - Miyuki Uno
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), Comprehensive Center for Precision Oncology (C2PO), Universidade de São Paulo, São Paulo 05508-220, Brazil; (R.C.); (M.U.); (D.D.A.)
| | - Diogo Dias Araújo
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), Comprehensive Center for Precision Oncology (C2PO), Universidade de São Paulo, São Paulo 05508-220, Brazil; (R.C.); (M.U.); (D.D.A.)
| | - Dalila Luciola Zanette
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81350-010, Brazil; (A.M.M.); (M.B.); (D.L.Z.)
| | - Mateus Nóbrega Aoki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81350-010, Brazil; (A.M.M.); (M.B.); (D.L.Z.)
| |
Collapse
|
11
|
YANG HONG, LI WAN, REN LIWEN, YANG YIHUI, ZHANG YIZHI, GE BINBIN, LI SHA, ZHENG XIANGJIN, LIU JINYI, ZHANG SEN, DU GUANHUA, TANG BO, WANG HONGQUAN, WANG JINHUA. Progress on diagnostic and prognostic markers of pancreatic cancer. Oncol Res 2023; 31:83-99. [PMID: 37304241 PMCID: PMC10208033 DOI: 10.32604/or.2023.028905] [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: 01/17/2023] [Accepted: 02/15/2023] [Indexed: 06/13/2023] Open
Abstract
Pancreatic cancer is a malignant disease characterized by low survival and high recurrence rate, whose patients are mostly at the stage of locally advanced or metastatic disease when first diagnosed. Early diagnosis is particularly important because prognostic/predictive markers help guide optimal individualized treatment regimens. So far, CA19-9 is the only biomarker for pancreatic cancer approved by the FDA, but its effectiveness is limited by low sensitivity and specificity. With recent advances in genomics, proteomics, metabolomics, and other analytical and sequencing technologies, the rapid acquisition and screening of biomarkers is now possible. Liquid biopsy also occupies a significant place due to its unique advantages. In this review, we systematically describe and evaluate the available biomarkers that have the greatest potential as vital tools in diagnosing and treating pancreatic cancer.
Collapse
Affiliation(s)
- HONG YANG
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - WAN LI
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - LIWEN REN
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - YIHUI YANG
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - YIZHI ZHANG
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - BINBIN GE
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - SHA LI
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - XIANGJIN ZHENG
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - JINYI LIU
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - SEN ZHANG
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - GUANHUA DU
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - BO TANG
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - HONGQUAN WANG
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - JINHUA WANG
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
12
|
Nené NR, Ney A, Nazarenko T, Blyuss O, Johnston HE, Whitwell HJ, Sedlak E, Gentry-Maharaj A, Apostolidou S, Costello E, Greenhalf W, Jacobs I, Menon U, Hsuan J, Pereira SP, Zaikin A, Timms JF. Serum biomarker-based early detection of pancreatic ductal adenocarcinomas with ensemble learning. COMMUNICATIONS MEDICINE 2023; 3:10. [PMID: 36670203 PMCID: PMC9860022 DOI: 10.1038/s43856-023-00237-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/04/2023] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Earlier detection of pancreatic ductal adenocarcinoma (PDAC) is key to improving patient outcomes, as it is mostly detected at advanced stages which are associated with poor survival. Developing non-invasive blood tests for early detection would be an important breakthrough. METHODS The primary objective of the work presented here is to use a dataset that is prospectively collected, to quantify a set of cancer-associated proteins and construct multi-marker models with the capacity to predict PDAC years before diagnosis. The data used is part of a nested case-control study within the UK Collaborative Trial of Ovarian Cancer Screening and is comprised of 218 samples, collected from a total of 143 post-menopausal women who were diagnosed with pancreatic cancer within 70 months after sample collection, and 249 matched non-cancer controls. We develop a stacked ensemble modelling technique to achieve robustness in predictions and, therefore, improve performance in newly collected datasets. RESULTS Here we show that with ensemble learning we can predict PDAC status with an AUC of 0.91 (95% CI 0.75-1.0), sensitivity of 92% (95% CI 0.54-1.0) at 90% specificity, up to 1 year prior to diagnosis, and at an AUC of 0.85 (95% CI 0.74-0.93) up to 2 years prior to diagnosis (sensitivity of 61%, 95% CI 0.17-0.83, at 90% specificity). CONCLUSIONS The ensemble modelling strategy explored here outperforms considerably biomarker combinations cited in the literature. Further developments in the selection of classifiers balancing performance and heterogeneity should further enhance the predictive capacity of the method.
Collapse
Affiliation(s)
- Nuno R Nené
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, 84-86 Chenies Mews, London, WC1E 6HU, UK.
- Institute for Women's Health, University College London, Cruciform Building 1.1, Gower Street, London, WC1E 6BT, UK.
| | - Alexander Ney
- Institute for Liver and Digestive Health, University College London, Upper 3rd Floor, Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Tatiana Nazarenko
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, 84-86 Chenies Mews, London, WC1E 6HU, UK
- Department of Mathematics, University College London, London, WC1H 0AY, UK
| | - Oleg Blyuss
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, 84-86 Chenies Mews, London, WC1E 6HU, UK
- Wolfson Institute of Population Health, Queen Mary University of London, Charterhouse Square, EC1M 6BQ, London, UK
| | - Harvey E Johnston
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, 84-86 Chenies Mews, London, WC1E 6HU, UK
- Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Harry J Whitwell
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, 84-86 Chenies Mews, London, WC1E 6HU, UK
- National Phenome Centre and Imperial Clinical Phenotyping Centre, Department of Metabolism, Digestion and Reproduction, IRDB Building, Imperial College London, Hammersmith Campus, London, W12 0NN, UK
- Section of Bioanalytical Chemistry, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Eva Sedlak
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, 84-86 Chenies Mews, London, WC1E 6HU, UK
| | - Aleksandra Gentry-Maharaj
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, 90 High Holborn, 2nd Floor, London, WC1V 6LJ, UK
| | - Sophia Apostolidou
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, 90 High Holborn, 2nd Floor, London, WC1V 6LJ, UK
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - William Greenhalf
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, L69 3GL, UK
| | - Ian Jacobs
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, 84-86 Chenies Mews, London, WC1E 6HU, UK
- University of New South Wales, Sydney, NSW, 2052, Australia
| | - Usha Menon
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, 90 High Holborn, 2nd Floor, London, WC1V 6LJ, UK
| | - Justin Hsuan
- Institute for Liver and Digestive Health, University College London, Upper 3rd Floor, Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Stephen P Pereira
- Institute for Liver and Digestive Health, University College London, Upper 3rd Floor, Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Alexey Zaikin
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, 84-86 Chenies Mews, London, WC1E 6HU, UK
- Department of Mathematics, University College London, London, WC1H 0AY, UK
| | - John F Timms
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, 84-86 Chenies Mews, London, WC1E 6HU, UK
| |
Collapse
|
13
|
Xu B, Chen Y, Peng M, Zheng JH, Zuo C. Exploring the potential of exosomes in diagnosis and drug delivery for pancreatic ductal adenocarcinoma. Int J Cancer 2023; 152:110-122. [PMID: 35765844 PMCID: PMC9796664 DOI: 10.1002/ijc.34195] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 05/30/2022] [Accepted: 06/09/2022] [Indexed: 01/07/2023]
Abstract
Pancreatic cancer (PC) is a cancer of the digestive system, and pancreatic ductal adenocarcinoma (PDAC) accounts for approximately 90% of all PC cases. Exosomes derived from PDAC (PDAC-exosomes) promote PDAC development and metastasis. Exosomes are nanoscale vesicles secreted by most cells, which can carry biologically active molecules and mediate communication and cargo transportation among cells. Recent studies have focused on transforming exosomes into good drug delivery systems (DDSs) to improve the clinical treatment of PDAC. This review considers PDAC as the main research object to introduce the role of PDAC-exosomes in PDAC development and metastasis. This review focuses on the following two themes: (a) the great potential of PDAC-exosomes as new diagnostic markers for PDAC, and (b) the transformation of exosomes into potential DDSs.
Collapse
Affiliation(s)
- Biaoming Xu
- Department of Gastroduodenal and Pancreatic SurgeryTranslational Medicine Joint Research Center of Liver Cancer of Hunan University, Laboratory of Digestive Oncology, Affiliated Cancer Hospital of Xiangya Medical School & Hunan Cancer Hospital, Central South UniversityChangshaChina
| | - Yu Chen
- Institute of Pathogen Biology and Immunology of College of BiologyHunan Provincial Key Laboratory of Medical Virology, Hunan UniversityChangshaChina
| | - Mingjing Peng
- Department of Gastroduodenal and Pancreatic SurgeryTranslational Medicine Joint Research Center of Liver Cancer of Hunan University, Laboratory of Digestive Oncology, Affiliated Cancer Hospital of Xiangya Medical School & Hunan Cancer Hospital, Central South UniversityChangshaChina
| | - Jin Hai Zheng
- Institute of Pathogen Biology and Immunology of College of BiologyHunan Provincial Key Laboratory of Medical Virology, Hunan UniversityChangshaChina
| | - Chaohui Zuo
- Department of Gastroduodenal and Pancreatic SurgeryTranslational Medicine Joint Research Center of Liver Cancer of Hunan University, Laboratory of Digestive Oncology, Affiliated Cancer Hospital of Xiangya Medical School & Hunan Cancer Hospital, Central South UniversityChangshaChina
| |
Collapse
|
14
|
Laser Capture Microdissection: A Gear for Pancreatic Cancer Research. Int J Mol Sci 2022; 23:ijms232314566. [PMID: 36498893 PMCID: PMC9741023 DOI: 10.3390/ijms232314566] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
The advancement in molecular techniques has been attributed to the quality and significance of cancer research. Pancreatic cancer (PC) is one of the rare cancers with aggressive behavior and a high mortality rate. The asymptomatic nature of the disease until its advanced stage has resulted in late diagnosis as well as poor prognosis. The heterogeneous character of PC has complicated cancer development and progression studies. The analysis of bulk tissues of the disease was insufficient to understand the disease, hence, the introduction of the single-cell separating technique aided researchers to decipher more about the specific cell population of tumors. This review gives an overview of the Laser Capture Microdissection (LCM) technique, one of the single-cell separation methods used in PC research.
Collapse
|
15
|
Balaban DV, Marin FS, Manucu G, Zoican A, Ciochina M, Mina V, Patoni C, Vladut C, Bucurica S, Costache RS, Ionita-Radu F, Jinga M. Clinical characteristics and outcomes in carbohydrate antigen 19-9 negative pancreatic cancer. World J Clin Oncol 2022; 13:630-640. [PMID: 36157158 PMCID: PMC9346420 DOI: 10.5306/wjco.v13.i7.630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/19/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of death from cancer worldwide. Tumor markers like carbohydrate antigen 19-9 (CA 19-9) have been proven valuable as a diagnostic tool and a predictor for tumor staging and response to therapy.
AIM To delineate the phenotype of normal CA 19-9 PDAC according to clinical features, disease staging and prognosis as compared with high CA 19-9 PDAC cases.
METHODS We performed a retrospective single-center analysis of all PDAC cases admitted in our Gastroenterology department over a period of 30 mo that were diagnosed by endoscopic ultrasound-guided tissue acquisition. Patients were divided into two groups according to CA 19-9 levels over a threshold of 37 U/mL. We performed a comparison between the two groups with regard to demographic and clinical data, biomarkers, tumor staging and 6-mo survival.
RESULTS Altogether 111 patients were recruited with 29 having documented normal CA 19-9 (< 37 U/mL). In the CA 19-9 negative group of patients, 20.68% had elevated levels of both CEA and CA 125, 13.79% for CA 125 only whilst 17.24% for CEA only. The two groups had similar demographic characteristics. Abdominal pain was more frequently reported in positive vs negative CA 19-9 PDAC cases (76.83% vs 55.17%), while smoking was slightly more prevalent in the latter group (28.04% vs 31.03%). Tumors over 2 cm were more frequently seen in the positive CA 19-9 group, reflecting a higher proportion of locally advanced and metastatic neoplasia (87.7% vs 79.3%). Six-month survival was higher for the negative CA 19-9 group (58.62% vs 47.56%).
CONCLUSION Elevated CA 19-9 at diagnosis seems to be associated with a more pronounced symptomatology, high tumor burden and poor prognosis compared to negative CA 19-9 PDAC cases. CEA and CA 125 can be adjunctive useful markers for PDAC, especially in CA 19-9 negative cases.
Collapse
Affiliation(s)
- Daniel Vasile Balaban
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Flavius Stefan Marin
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
- Department of Gastroenterology and Digestive Oncology, Hôpital Cochin, Paris 75014, France
| | - George Manucu
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Andreea Zoican
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Marina Ciochina
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Victor Mina
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Cristina Patoni
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Catalina Vladut
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Prof Dr. Agrippa Ionescu Clinical Emergency Hospital, Bucharest 020021, Romania
| | - Sandica Bucurica
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Raluca Simona Costache
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Florentina Ionita-Radu
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| | - Mariana Jinga
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest 020021, Romania
| |
Collapse
|
16
|
Clinicopathological and Prognostic Value of Survivin Expression in Surgically Resected Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14143494. [PMID: 35884555 PMCID: PMC9322386 DOI: 10.3390/cancers14143494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/10/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Survival after surgery for pancreatic ductal adenocarcinoma (PDAC) remains poor. Thus, novel therapeutic concepts focus on the development of targeted therapies. In this context, inhibitor of apoptosis protein (IAP) survivin is regarded as a promising oncotherapeutic target. However, its expression and prognostic value in different tumour compartments of PDAC have not been studied. Methods: Immunohistochemical analysis of survivin in different PDAC tumour compartments from 236 consecutive patients was correlated with clinicopathological variables and survival. Results: In comparison to healthy pancreatic tissue high nuclear (p < 0.001) and high cytoplasmic (p < 0.01) survivin expression became evident in the tumour centre, along the invasion front and in lymph node metastases. Cytoplasmic overexpression of survivin in tumour centres was related to the presence of distant metastasis (p = 0.016) and UICC III/IV stages (p = 0.009), while high cytoplasmic expression at the invasion front grouped with venous infiltration (p = 0.022). Increased nuclear survivin along the invasion front correlated with perineural invasion (p = 0.035). High nuclear survivin in tumour centres represented an independent prognostic factor for overall survival of pancreatic tail carcinomas (HR 13.5 95%CI (1.4−129.7)) and correlated with a limited disease-free survival in PDAC (HR 1.80 95%CI (1.04−3.12)). Conclusion: Survivin is associated with advanced disease stages and poor prognosis. Therefore, survivin will help to identify patients with aggressive tumour phenotypes that could benefit from the inclusion in clinical trials incorporating survivin inhibitors in PDAC.
Collapse
|
17
|
Li W, Li T, Sun C, Du Y, Chen L, Du C, Shi J, Wang W. Identification and prognostic analysis of biomarkers to predict the progression of pancreatic cancer patients. Mol Med 2022; 28:43. [PMID: 35428170 PMCID: PMC9013045 DOI: 10.1186/s10020-022-00467-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/04/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is a malignancy with a poor prognosis and high mortality. Surgical resection is the only "curative" treatment. However, only a minority of patients with PC can obtain surgery. Improving the overall survival (OS) rate of patients with PC is still a major challenge. Molecular biomarkers are a significant approach for diagnostic and predictive use in PCs. Several prediction models have been developed for patients newly diagnosed with PC that is operable or patients with advanced and metastatic PC; however, these models require further validation. Therefore, precise biomarkers are urgently required to increase the efficiency of predicting a disease-free survival (DFS), OS, and sensitivity to immunotherapy in PC patients and to improve the prognosis of PC. METHODS In the present study, we first evaluated the highly and selectively expressed targets in PC, using the GeoMxTM Digital Spatial Profiler (DSP) and then, we analyzed the roles of these targets in PCs using TCGA database. RESULTS LAMB3, FN1, KRT17, KRT19, and ANXA1 were defined as the top five upregulated targets in PC compared with paracancer. The TCGA database results confirmed the expression pattern of LAMB3, FN1, KRT17, KRT19, and ANXA1 in PCs. Significantly, LAMB3, FN1, KRT19, and ANXA1 but not KRT17 can be considered as biomarkers for survival analysis, univariate and multivariate Cox proportional hazards model, and risk model analysis. Furthermore, in combination, LAMB3, FN1, KRT19, and ANXA1 predict the DFS and, in combination, LAMB3, KRT19, and ANXA1 predict the OS. Immunotherapy is significant for PCs that are inoperable. The immune checkpoint blockade (ICB) analysis indicated that higher expressions of FN1 or ANXA1 are correlated with lower ICB response. In contrast, there are no significant differences in the ICB response between high and low expression of LAMB3 and KRT19. CONCLUSIONS In conclusion, LAMB3, FN1, KRT19, and ANXA1 are good predictors of PC prognosis. Furthermore, FN1 and ANXA1 can be predictors of immunotherapy in PCs.
Collapse
Affiliation(s)
- Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- The Academy of Medical Science, College of Medical, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Tiandong Li
- College of Public Health, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chenguang Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yimeng Du
- The Academy of Medical Science, College of Medical, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Linna Chen
- The Academy of Medical Science, College of Medical, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chunyan Du
- Laboratory Animal Center, School of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Jianxiang Shi
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Science, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Weijie Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
18
|
Mortoglou M, Buha Djordjevic A, Djordjevic V, Collins H, York L, Mani K, Valle E, Wallace D, Uysal-Onganer P. Role of microRNAs in response to cadmium chloride in pancreatic ductal adenocarcinoma. Arch Toxicol 2022; 96:467-485. [PMID: 34905088 PMCID: PMC8837568 DOI: 10.1007/s00204-021-03196-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/10/2021] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most fatal and aggressive malignancies with a 5-year survival rate less than 9%. Early detection is particularly difficult due to the lack of symptoms even in advanced stages. microRNAs (miRs/miRNAs) are small (~ 18-24 nucleotides), endogenous, non-coding RNAs, which are involved in the pathogenesis of several malignancies including PDAC. Alterations of miR expressions can lead to apoptosis, angiogenesis, and metastasis. The role of environmental pollutants such as cadmium (Cd) in PDAC has been suggested but not fully understood. This study underlines the role of miRs (miR-221, miR-155, miR-126) in response to cadmium chloride (CdCl2) in vitro. Lethal concentration (LC50) values for CdCl2 resulted in a toxicity series of AsPC-1 > HPNE > BxPC-3 > Panc-1 = Panc-10.5. Following the treatment with CdCl2, miR-221 and miR-155 were significantly overexpressed, whereas miR-126 was downregulated. An increase in epithelial-mesenchymal transition (EMT) via the dysregulation of mesenchymal markers such as Wnt-11, E-cadherin, Snail, and Zeb1 was also observed. Hence, this study has provided evidence to suggest that the environmental pollutant Cd can have a significant role in the development of PDAC, suggesting a significant correlation between miRs and Cd exposure during PDAC progression. Further studies are needed to investigate the precise role of miRs in PDAC progression as well as the role of Cd and other environmental pollutants.
Collapse
Affiliation(s)
- Maria Mortoglou
- Cancer Research Group, School of Life Sciences, University of Westminster, London, W1W 6UW UK
| | | | | | - Hunter Collins
- College of Medicine and the Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, 1111 West 17th Street, Tulsa, OK 74107-1898 USA
| | - Lauren York
- College of Medicine and the Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, 1111 West 17th Street, Tulsa, OK 74107-1898 USA
| | - Katherine Mani
- College of Medicine and the Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, 1111 West 17th Street, Tulsa, OK 74107-1898 USA
| | - Elizabeth Valle
- College of Medicine and the Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, 1111 West 17th Street, Tulsa, OK 74107-1898 USA
| | - David Wallace
- College of Medicine and the Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, 1111 West 17th Street, Tulsa, OK 74107-1898 USA
| | - Pinar Uysal-Onganer
- Cancer Research Group, School of Life Sciences, University of Westminster, London, W1W 6UW UK
| |
Collapse
|
19
|
Song J, Sokoll LJ, Chan DW, Zhang Z. Validation of Serum Biomarkers That Complement CA19-9 in Detecting Early Pancreatic Cancer Using Electrochemiluminescent-Based Multiplex Immunoassays. Biomedicines 2021; 9:biomedicines9121897. [PMID: 34944713 PMCID: PMC8698985 DOI: 10.3390/biomedicines9121897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy; its early detection is critical for improving prognosis. Electrochemiluminescent-based multiplex immunoassays were developed with high analytical performance. All proteins were analyzed in sera of patients diagnosed with PDAC (n = 138), benign pancreatic conditions (111), and healthy controls (70). The clinical performance of these markers was evaluated individually or in combination for their complementarity to CA19-9 in detecting early PDAC. Logistic regression modeling including sex and age as cofactors identified a two-marker panel of CA19-9 and CA-125 that significantly improved the performance of CA19-9 alone in discriminating PDAC (AUC: 0.857 vs. 0.766), as well as early stage PDAC (0.805 vs. 0.702) from intraductal papillary mucinous neoplasm (IPMN). At a fixed specificity of 80%, the panel significantly improved sensitivities (78% vs. 41% or 72% vs. 59%). A two-marker panel of HE4 and CEA significantly outperformed CA19-9 in separating IPMN from chronic pancreatitis (0.841 vs. 0.501). The biomarker panels evaluated by assays demonstrated potential complementarity to CA19-9 in detecting early PDAC, warranting additional clinical validation to determine their role in the early detection of pancreatic cancer.
Collapse
Affiliation(s)
- Jin Song
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, 419 North Caroline Street, Baltimore, MD 21231, USA; (L.J.S.); (D.W.C.)
- Correspondence: (J.S.); (Z.Z.); Tel.: +1-443-287-6363 (J.S.); +1-410-502-7871 (Z.Z.)
| | - Lori J. Sokoll
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, 419 North Caroline Street, Baltimore, MD 21231, USA; (L.J.S.); (D.W.C.)
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Daniel W. Chan
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, 419 North Caroline Street, Baltimore, MD 21231, USA; (L.J.S.); (D.W.C.)
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Zhen Zhang
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, 419 North Caroline Street, Baltimore, MD 21231, USA; (L.J.S.); (D.W.C.)
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Correspondence: (J.S.); (Z.Z.); Tel.: +1-443-287-6363 (J.S.); +1-410-502-7871 (Z.Z.)
| |
Collapse
|
20
|
Kapszewicz M, Małecka-Wojciesko E. Simple Serum Pancreatic Ductal Adenocarcinoma (PDAC) Protein Biomarkers-Is There Anything in Sight? J Clin Med 2021; 10:jcm10225463. [PMID: 34830745 PMCID: PMC8619303 DOI: 10.3390/jcm10225463] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/07/2021] [Accepted: 11/20/2021] [Indexed: 01/04/2023] Open
Abstract
A poor PDAC prognosis is due to a lack of effective treatment and late diagnosis. The early detection of PDAC could significantly decrease mortality and save lives. Idealbiomarkers for PDAC should be cost-effective, detectable in easily accessible biological material, and present in sufficient concentration in the earliest possible phase of the disease. This review addresses newly selected, simple protein biomarkers—new ones such as thrombospondin-2, insulin-linked binding protein 2, lysophosphatidic acid, and autotaxin and conventional ones such as Ca19-9, inflammatory factors, and coagulation factors. Their possible use in the early detection of PDAC, differentiation from benign diseases, prognosis, and treatment response prediction is discussed. We also address the usefulness of possible combinations of biomarkers in diagnostic panels.
Collapse
|
21
|
Umans DS, Hoogenboom SA, Sissingh NJ, Lekkerkerker SJ, Verdonk RC, van Hooft JE. Pancreatitis and pancreatic cancer: A case of the chicken or the egg. World J Gastroenterol 2021; 27:3148-3157. [PMID: 34163103 PMCID: PMC8218365 DOI: 10.3748/wjg.v27.i23.3148] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/13/2021] [Accepted: 05/08/2021] [Indexed: 02/06/2023] Open
Abstract
Acute pancreatitis (AP), chronic pancreatitis (CP) and pancreatic cancer are three distinct pancreatic diseases with different prognoses and treatment options. However, it may be difficult to differentiate between benign and malignant disease. AP may be a first symptom of pancreatic cancer, particularly in patients between the ages of 56 and 75 with presumed idiopathic AP who had a concomitant diagnosis of new-onset diabetes mellitus or patients who present with CP at diagnosis of AP. In these patients, additional imaging is warranted, preferably by endoscopic ultrasonography. CP may lead to pancreatic cancer through oncogenic mutations, mostly in patients with hereditary CP, and in patients in whom risk factors for pancreatic cancer (e.g., nicotine and alcohol abuse) are also present. Patients with PRSS1-mediated CP and patients with a history of autosomal dominant hereditary CP without known genetic mutations may be considered for surveillance for pancreatic cancer. Pancreatic inflammation may mimic pancreatic cancer by appearing as a focal mass-forming lesion on imaging. Differentiation between the above mentioned benign and malignant disease may be facilitated by specific features like the duct-penetrating sign and the duct-to-parenchyma ratio. Research efforts are aimed towards developing a superior discriminant between pancreatitis and pancreatic cancer in the form of imaging modalities or biomarkers. This may aid clinicians in timely diagnosing pancreatic cancer in a potentially curable stage.
Collapse
Affiliation(s)
- Devica S Umans
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology and Metabolism, Amsterdam UMC, Amsterdam 1105 AZ, The Netherlands
- Department of Research and Development, St. Antonius Hospital, Nieuwegein 3430 EM, The Netherlands
| | - Sanne A Hoogenboom
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology and Metabolism, Amsterdam UMC, Amsterdam 1105 AZ, The Netherlands
| | - Noor J Sissingh
- Department of Research and Development, St. Antonius Hospital, Nieuwegein 3430 EM, The Netherlands
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Selma J Lekkerkerker
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology and Metabolism, Amsterdam UMC, Amsterdam 1105 AZ, The Netherlands
| | - Robert C Verdonk
- Department of Gastroenterology and Hepatology, St. Antonius Hospital, Nieuwegein 3430 EM, The Netherlands
| | - Jeanin E van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| |
Collapse
|
22
|
Turanli B, Yildirim E, Gulfidan G, Arga KY, Sinha R. Current State of "Omics" Biomarkers in Pancreatic Cancer. J Pers Med 2021; 11:127. [PMID: 33672926 PMCID: PMC7918884 DOI: 10.3390/jpm11020127] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the most fatal malignancies and the seventh leading cause of cancer-related deaths related to late diagnosis, poor survival rates, and high incidence of metastasis. Unfortunately, pancreatic cancer is predicted to become the third leading cause of cancer deaths in the future. Therefore, diagnosis at the early stages of pancreatic cancer for initial diagnosis or postoperative recurrence is a great challenge, as well as predicting prognosis precisely in the context of biomarker discovery. From the personalized medicine perspective, the lack of molecular biomarkers for patient selection confines tailored therapy options, including selecting drugs and their doses or even diet. Currently, there is no standardized pancreatic cancer screening strategy using molecular biomarkers, but CA19-9 is the most well known marker for the detection of pancreatic cancer. In contrast, recent innovations in high-throughput techniques have enabled the discovery of specific biomarkers of cancers using genomics, transcriptomics, proteomics, metabolomics, glycomics, and metagenomics. Panels combining CA19-9 with other novel biomarkers from different "omics" levels might represent an ideal strategy for the early detection of pancreatic cancer. The systems biology approach may shed a light on biomarker identification of pancreatic cancer by integrating multi-omics approaches. In this review, we provide background information on the current state of pancreatic cancer biomarkers from multi-omics stages. Furthermore, we conclude this review on how multi-omics data may reveal new biomarkers to be used for personalized medicine in the future.
Collapse
Affiliation(s)
- Beste Turanli
- Department of Bioengineering, Marmara University, 34722 Istanbul, Turkey; (B.T.); (E.Y.); (G.G.)
| | - Esra Yildirim
- Department of Bioengineering, Marmara University, 34722 Istanbul, Turkey; (B.T.); (E.Y.); (G.G.)
| | - Gizem Gulfidan
- Department of Bioengineering, Marmara University, 34722 Istanbul, Turkey; (B.T.); (E.Y.); (G.G.)
| | - Kazim Yalcin Arga
- Department of Bioengineering, Marmara University, 34722 Istanbul, Turkey; (B.T.); (E.Y.); (G.G.)
- Turkish Institute of Public Health and Chronic Diseases, 34718 Istanbul, Turkey
| | - Raghu Sinha
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
23
|
Chen J, Li H, Xu W, Guo X. Evaluation of serum ATX and LPA as potential diagnostic biomarkers in patients with pancreatic cancer. BMC Gastroenterol 2021; 21:58. [PMID: 33568105 PMCID: PMC7877052 DOI: 10.1186/s12876-021-01635-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 02/02/2021] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is a devastating disease that has a poor prognosis and a total 5-year survival rate of around 5%. The poor prognosis of PC is due in part to a lack of suitable biomarkers that can allow early diagnosis. The lysophospholipase autotaxin (ATX) and its product lysophosphatidic acid (LPA) play an essential role in disease progression in PC patients and are associated with increased morbidity in several types of cancer. In this study, we evaluated both the potential role of serum LPA and ATX as diagnostic markers in PC and their prognostic value for PC either alone or in combination with CA19-9. METHODS ATX, LPA and CA19-9 levels were evaluated using ELISA of serum obtained from PC patients (n = 114) healthy volunteers (HVs: n = 120) and patients with benign pancreatic diseases (BPDs: n = 94). RESULTS Serum levels of ATX, LPA and CA19-9 in PC patients were substantially higher than that for BPD patients or HVs (p < 0.001). The sensitivity of LPA in early phase PC was 91.74% and the specificity of ATX was 80%. The levels of ATX, LPA and CA19-9 were all substantially higher for early stage PC patients compared to levels in serum from BPD patients and HVs. The diagnostic efficacy of CA19-9 for PC was significantly enhanced by the addition of ATX and LPA (p = 0.0012). CONCLUSION Measurement of LPA and ATX levels together with CA19-9 levels can be used for early detection of PC and diagnosis of PC in general.
Collapse
Affiliation(s)
- Jiang Chen
- Department of Gastroenterology, General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenyang, 110840, Liaoning Province, China
| | - Hongyu Li
- Department of Gastroenterology, General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenyang, 110840, Liaoning Province, China
| | - Wenda Xu
- Department of Gastroenterology, General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenyang, 110840, Liaoning Province, China
| | - Xiaozhong Guo
- Department of Gastroenterology, General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenyang, 110840, Liaoning Province, China.
| |
Collapse
|
24
|
Abstract
OBJECTIVES Pancreatic cancer (PC) is a highly malignant tumor with poor detection sensitivity and specificity in biomarkers and diagnosis. Previous research indicated that serum Ephrin type-A receptor 2 in exosomes (Exo-EphA2) was highly expressed and might have facilitated cell migration in PC cells. However, the dynamics of clinical performance of serum Exo-EphA2 in PC patients are unknown. Thus, this study evaluated serum Exo-EphA2 as a potential diagnostic biomarker in PC. METHODS The expressions of serum Exo-EphA2 were assessed by enzyme-linked immunosorbent assay for N = 353 serum samples, including from 204 PC patients, 75 patients with benign pancreatic disease, and 74 healthy control patients. Carbohydrate antigen 19-9 (CA 19-9) and carbohydrate antigen 242 (CA 242) were measured by automated immunoassay. RESULTS Serum Exo-EphA2 levels were significantly higher in PC patients than in benign pancreatic disease and healthy control patients. Receiver operating characteristic curve analysis suggested that using combined diagnoses of Exo-EphA2 with CA 19-9 and CA 242 was more effective to discriminate early stage (stage I and II) in PC than in healthy controls and benign disease patients. CONCLUSIONS Novel findings suggest that serum Exo-EphA2 is a potential early diagnostic biomarker complementing CA 19-9 and CA 242 in PC.
Collapse
|
25
|
Khatri I, Bhasin MK. A Transcriptomics-Based Meta-Analysis Combined With Machine Learning Identifies a Secretory Biomarker Panel for Diagnosis of Pancreatic Adenocarcinoma. Front Genet 2020; 11:572284. [PMID: 33133160 PMCID: PMC7511758 DOI: 10.3389/fgene.2020.572284] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/12/2020] [Indexed: 12/22/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is generally incurable due to the late diagnosis and absence of markers that are concordant with expression in several sample sources (i.e., tissue, blood, plasma) and platforms (i.e., Microarray, sequencing). We optimized meta-analysis of 19 PDAC (tissue and blood) transcriptome studies from multiple platforms. The key biomarkers for PDAC diagnosis with secretory potential were identified and validated in different cohorts. Machine learning approach i.e., support vector machine supported by leave-one-out cross-validation was used to build and test the classifier. We identified a 9-gene panel (IFI27, ITGB5, CTSD, EFNA4, GGH, PLBD1, HTATIP2, IL1R2, CTSA) that achieved ∼0.92 average sensitivity and ∼0.90 average specificity in distinguishing PDAC from healthy samples in five training sets using cross-validation. These markers were also validated in proteomics and single-cell transcriptomics studies suggesting their prognostic role in the diagnosis of PDAC. Our 9-gene classifier can not only clearly discriminate between better and poor survivors but can also precisely discriminate PDAC from chronic pancreatitis (AUC = 0.95), early stages of progression [Stage I and II (AUC = 0.82), IPMA and IPMN (AUC = 1), and IPMC (AUC = 0.81)]. The 9-gene marker outperformed the previously known markers in blood studies particularly (AUC = 0.84). The discrimination of PDAC from early precursor lesions in non-malignant tissue (AUC > 0.81) and peripheral blood (AUC > 0.80) may assist in an early diagnosis of PDAC in blood samples and thus will also facilitate risk stratification upon validation in clinical trials.
Collapse
Affiliation(s)
- Indu Khatri
- Division of IMBIO, Department of Medicine, Beth Israel Lahey Health, Harvard Medical School, Boston, MA, United States.,Department of Immunology and Leiden Computational Biology Center, Leiden University Medical Center, Leiden, Netherlands
| | - Manoj K Bhasin
- Division of IMBIO, Department of Medicine, Beth Israel Lahey Health, Harvard Medical School, Boston, MA, United States.,Department of Pediatrics and Biomedical Informatics, Children's Healthcare of Atlanta, Emory School of Medicine, Atlanta, GA, United States
| |
Collapse
|
26
|
Ariston Gabriel AN, Wang F, Jiao Q, Yvette U, Yang X, Al-Ameri SA, Du L, Wang YS, Wang C. The involvement of exosomes in the diagnosis and treatment of pancreatic cancer. Mol Cancer 2020; 19:132. [PMID: 32854710 PMCID: PMC7450552 DOI: 10.1186/s12943-020-01245-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 08/12/2020] [Indexed: 12/24/2022] Open
Abstract
At the moment, pancreatic cancer is among the deadliest gastrointestinal diseases, and pancreatic cancer growth is a complex biological process that is based on different kinds of genes. Exosomes are extracellular vesicles containing microRNAs (miRNAs), messenger RNA (mRNA), and proteins, they act as the most prominent mediator of intercellular communication, and they regulate, instruct, and re-educate their surrounding microenvironment and target specific organs. Due to accumulative evidence proved that exosomes are involved in metastasis, cell proliferation, EMT, angiogenesis, and TME of pancreatic cancer, exosomes are crucial potential candidates to detect pancreatic cancer early. This review aims to convey the current understanding of the main functions employed by exosomes in early diagnosis and treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Abakundana Nsenga Ariston Gabriel
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, People's Republic of China
- Department of Clinical Laboratory Diagnostics, School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Fang Wang
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, People's Republic of China
| | - Qinlian Jiao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, People's Republic of China
- Marine College, Shandong University, Weihai, 264209, People's Republic of China
| | - Umwali Yvette
- Department of Clinical Laboratory Diagnostics, School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People's Republic of China
- Department of Clinical Laboratory, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Xuemei Yang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, People's Republic of China
| | - Samed Ahmed Al-Ameri
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, People's Republic of China
- Department of Clinical Laboratory Diagnostics, School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, People's Republic of China.
| | - Yun-Shan Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, People's Republic of China.
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, People's Republic of China.
| |
Collapse
|
27
|
Lin S, Fang Y, Mo Z, Lin Y, Ji C, Jian Z. Prognostic value of lymphocyte to monocyte ratio in pancreatic cancer: a systematic review and meta-analysis including 3338 patients. World J Surg Oncol 2020; 18:186. [PMID: 32711514 PMCID: PMC7382838 DOI: 10.1186/s12957-020-01962-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 07/17/2020] [Indexed: 01/09/2023] Open
Abstract
Background Recently, reports have classified lymphocyte to monocyte ratio (LMR) as an effective indicator for predicting the prognosis of pancreatic cancer. Nevertheless, the prognostic value of LMR for pancreatic cancer remains controversial. Through meta-analysis, this work intends to evaluate the potential prognostic role of pretreatment LMR in patients diagnosed with pancreatic cancer. Methods We reviewed and extracted eligible articles from Web of Science, PubMed, Cochrane Library, and Embase. A meta-analysis was conducted using hazard ratio (HR) and 95% confidence intervals (CIs) to assess the comparison between pretreatment LMR and overall survival (OS) and disease-free survival/recurrence-free survival/time to progression (DFS/RFS/TTP). Results In total, 11 studies (16 cohorts) including 3338 patients diagnosed with pancreatic cancer (PC) were enrolled in our meta-analysis. Notably, we revealed that high pretreatment LMR predicted better overall survival (OS) (HR = 0.68, 95% CI 0.58–0.80, P < 0.001, I-squared = 69.3%, Ph < 0.001) and DFS/RFS/TTP (HR = 0.55, 95% CI 0.31–0.96, P = 0.037, I-squared = 89.9%, Ph < 0.001) in patients with pancreatic cancer. Further, through subgroup analyses, we showed that high pretreatment LMR was significantly associated with the favorable OS regardless of ethnicity, study design, treatment method, variable type, the cut-off value for LMR, and disease stages of I–IV and III–IV. Conclusion The findings from our study suggest that high pretreatment LMR is associated with better OS and DFS/RFS/TTP in patients diagnosed with pancreatic cancer. As such, it can potentially serve as a novel prognostic biomarker for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Shuwen Lin
- Department of General Surgery, Binhaiwan Central Hospital of Dongguan (also called The Fifth People's Hospital of Dongguan), The Dongguan Affiliated Hospital of Medical College of Jinan University, Dongguan, 523905, Guangdong, People's Republic of China
| | - Yinghua Fang
- Department of Pain, Binhaiwan Central Hospital of Dongguan (also called The Fifth People's Hospital of Dongguan), The Dongguan Affiliated Hospital of Medical College of Jinan University, Dongguan, 523905, Guangdong, People's Republic of China
| | - Zhikang Mo
- Department of General Surgery, Binhaiwan Central Hospital of Dongguan (also called The Fifth People's Hospital of Dongguan), The Dongguan Affiliated Hospital of Medical College of Jinan University, Dongguan, 523905, Guangdong, People's Republic of China
| | - Ye Lin
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106 Zhongshan Er Road, Yuexiu, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Chenggang Ji
- Department of General Surgery, Binhaiwan Central Hospital of Dongguan (also called The Fifth People's Hospital of Dongguan), The Dongguan Affiliated Hospital of Medical College of Jinan University, Dongguan, 523905, Guangdong, People's Republic of China
| | - Zhixiang Jian
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106 Zhongshan Er Road, Yuexiu, Guangzhou, 510080, Guangdong, People's Republic of China.
| |
Collapse
|
28
|
Flammang I, Reese M, Yang Z, Eble JA, Dhayat SA. Tumor-Suppressive miR-192-5p Has Prognostic Value in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2020; 12:E1693. [PMID: 32630552 PMCID: PMC7352756 DOI: 10.3390/cancers12061693] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by fast tumor progression and diagnosis at advanced, inoperable stages. Previous studies could demonstrate an involvement of miR-192-5p in epigenetic regulation of visceral carcinomas. Due to contradictory results, however, the clinical utility of miR-192-5p in PDAC has yet to be determined. MiR-192-5p expression was analyzed by RT-qRT-PCR in human PDAC and benign tissue (n = 78), blood serum (n = 81) and serum exosomes (n = 74), as well as in PDAC cell lines (n = 5), chemoresistant cell clones (n = 2), and pancreatic duct cell line H6c7. Analysis of EMT-associated (epithelial-to-mesenchymal transition) proteins was performed by immunohistochemistry and Western blot. MiR-192-5p was deregulated in PDAC as compared to healthy controls (HCs), with downregulation in macrodissected tissue (p < 0.001) and upregulation in blood serum of PDAC UICC (Union for International Cancer Control) stage IV (p = 0.016) and serum exosomes of PDAC UICC stages II to IV (p < 0.001). MiR-192-5p expression in tumor tissue was significantly lower as compared to corresponding peritumoral tissue (PDAC UICC stage II: p < 0.001; PDAC UICC stage III: p = 0.024), while EMT markers ZEB1 and ZEB2 were more frequently expressed in tumor tissue as compared to peritumoral tissue, HCs, and chronic pancreatitis. Tissue-derived (AUC of 0.86; p < 0.0001) and exosomal (AUC of 0.83; p = 0.0004) miR-192-5p could differentiate between PDAC and HCs with good accuracy. Furthermore, high expression of miR-192-5p in PDAC tissue of curatively resected PDAC patients correlated with prolonged overall and recurrence-free survival in multivariate analysis. In vitro, miR-192-5p was downregulated in gemcitabine-resistant cell clones of AsPC-1 (p = 0.029). Transient transfection of MIA PaCa-2 cells with miR-192-5p mimic resulted in downregulation of ZEB2. MiR-192-5p seems to possess a tumor-suppressive role and high potential as a diagnostic and prognostic marker in PDAC.
Collapse
Affiliation(s)
- Isabelle Flammang
- Department of General, Visceral and Transplantation Surgery, University Hospital Muenster, Albert-Schweitzer-Campus 1 (W1), 48149 Muenster, Germany; (I.F.); (M.R.); (Z.Y.)
| | - Moritz Reese
- Department of General, Visceral and Transplantation Surgery, University Hospital Muenster, Albert-Schweitzer-Campus 1 (W1), 48149 Muenster, Germany; (I.F.); (M.R.); (Z.Y.)
| | - Zixuan Yang
- Department of General, Visceral and Transplantation Surgery, University Hospital Muenster, Albert-Schweitzer-Campus 1 (W1), 48149 Muenster, Germany; (I.F.); (M.R.); (Z.Y.)
| | - Johannes A. Eble
- Department of Physiological Chemistry and Pathobiochemistry, University of Muenster, Waldeyerstrasse 15, 48149 Muenster, Germany;
| | - Sameer A. Dhayat
- Department of General, Visceral and Transplantation Surgery, University Hospital Muenster, Albert-Schweitzer-Campus 1 (W1), 48149 Muenster, Germany; (I.F.); (M.R.); (Z.Y.)
| |
Collapse
|
29
|
Wang XY, Chen HT, Na R, Jiang DK, Lin XL, Yang F, Jin C, Fu DL, Xu JF. Single-nucleotide polymorphisms based genetic risk score in the prediction of pancreatic cancer risk. World J Gastroenterol 2020; 26:3076-3086. [PMID: 32587449 PMCID: PMC7304113 DOI: 10.3748/wjg.v26.i22.3076] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/29/2020] [Accepted: 05/12/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Disease-related single nucleotide polymorphisms (SNPs) based genetic risk score (GRS) has been proven to provide independent inherited risk other than family history in multiple cancer types.
AIM To evaluate the potential of GRS in the prediction of pancreatic cancer risk.
METHODS In this case-control study (254 cases and 1200 controls), we aimed to evaluate the association between GRS and pancreatic ductal adenocarcinoma (PDAC) risk in the Chinese population. The GRS was calculated based on the genotype information of 18 PDAC-related SNPs for each study subject (personal genotyping information of the SNPs) and was weighted by external odd ratios (ORs).
RESULTS GRS was significantly different in cases and controls (1.96 ± 3.84 in PDACs vs 1.09 ± 0.94 in controls, P < 0.0001). Logistic regression revealed GRS to be associated with PDAC risk [OR = 1.23, 95% confidence interval (CI): 1.13-1.34, P < 0.0001]. GRS remained significantly associated with PDAC (OR = 1.36, 95%CI: 1.06-1.74, P = 0.015) after adjusting for age and sex. Further analysis revealed an association of increased risk for PDAC with higher GRS. Compared with low GRS (< 1.0), subjects with high GRS (2.0) were 99% more likely to have PDAC (OR: 1.99, 95%CI: 1.30-3.04, P = 0.002). Participants with intermediate GRS (1.0-1.9) were 39% more likely to have PDAC (OR: 1.39, 95%CI: 1.03-1.84, P = 0.031). A positive trend was observed (P trend = 0.0006).
CONCLUSION GRS based on PDAC-associated SNPs could provide independent information on PDAC risk and may be used to predict a high risk PDAC population.
Collapse
Affiliation(s)
- Xiao-Yi Wang
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Hai-Tao Chen
- State Key Laboratory of Organ Failure Research, Guangdong Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Rong Na
- Shanghai Medical College, Fudan University, Shanghai 200043, China
- Department of Urology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - De-Ke Jiang
- State Key Laboratory of Organ Failure Research, Guangdong Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Xiao-Ling Lin
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai 200433, China
| | - Feng Yang
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chen Jin
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - De-Liang Fu
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jian-Feng Xu
- Program for Personalized Cancer Care and Department of Surgery, North Shore University Health System, Evanston, IL 60201, United States
| |
Collapse
|
30
|
Zhang J, Wang Y, Zhao T, Li Y, Tian L, Zhao J, Zhang J. Evaluation of serum MUC5AC in combination with CA19-9 for the diagnosis of pancreatic cancer. World J Surg Oncol 2020; 18:31. [PMID: 32028958 PMCID: PMC7006398 DOI: 10.1186/s12957-020-1809-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 01/28/2020] [Indexed: 12/15/2022] Open
Abstract
Background Pancreatic cancer (PC) is a highly aggressive tumor with a poor prognosis that lacks specific diagnostic markers. Mucin 5AC (MUC5AC) is a member of the mucin family, a heterogeneous group of high molecular weight, heavily glycosylated proteins that could be either membrane-bound or secreted. This multi-central study is to evaluate the performance of serum MUC5AC in combination with carbohydrate antigen 19-9 (CA19-9) for the diagnosis of PC in Asian. Methods Sixty-one patients with PC (comprised of early pancreatic cancer [n = 30] and late pancreatic cancer [n = 31] patients), 29 benign control, 35 choledocholithiasis, 25 chronic pancreatitis, and 34 healthy controls, were recruited from two hospitals. Serum levels of MUC5AC were evaluated by commercial ELISA kits. CA19-9 was measured by chemiluminescence immunoassay. The cutoff value of MUC5AC was determined based on optimal sensitivity and specificity. Results Serum MUC5AC in patients with PC (210.1 [100.5–423.8] ng/mL) presented higher levels than those in controls. The combined biomarker panel (MUC5AC and CA19-9) presented better performance and improved specificity to differentiate PC from controls (AUC 0.894; 95% CI (0.844–0.943), sensitivity 0.738, specificity 0.886) than CA19-9 (p = 0.043) or MUC5AC alone (p = 0.010); however, the latter two had no difference (p = 0.824). Conclusions Serum MUC5AC is a potential biomarker for PC. The combination with CA19-9 presents improved specificity and better performance.
Collapse
Affiliation(s)
- Jiayu Zhang
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yue Wang
- Department of Hepatology, The Fifth People's Hospital of Suzhou, Suzhou, China
| | - Tiancheng Zhao
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Yezhou Li
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Leilei Tian
- Operating Room, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinming Zhao
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jingxin Zhang
- Department of General Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
31
|
Design of an immunohistochemistry biomarker panel for diagnosis of pancreatic adenocarcinoma. Pancreatology 2019; 19:842-849. [PMID: 31445888 DOI: 10.1016/j.pan.2019.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Challenges still exist in differentiating pancreatic adenocarcinoma from benign disease. The use of adjuvant testing of tissue biopsies has demonstrated potential diagnostic value. We designed a proof of concept study to first validate four individual immunohistochemistry biomarkers and then combine them into a panel to boost overall diagnostic sensitivity. METHODS Malignant and benign pancreas from 27 pancreaticoduodenectomy specimens underwent immunohistochemistry staining with VHL, IMP3, S100A4, S100P. Using ROC curve analysis, threshold criteria for number of cells staining were chosen for each biomarker. Biomarkers were then evaluated as a panel for their ability to discriminate malignant from benign specimens. RESULTS Diagnostic sensitivity of VHL, IMP3, S100A4, and S100P were 75.0%, 79.2%, 45.8%, and 0%. When VHL, IMP3, and S100A4 were grouped into a panel, they were able to distinguish cancer from normal tissue with a sensitivity of 100% and a specificity of 96%. CONCLUSIONS The high diagnostic value of an IHC panel consisting of VHL, IMP3, and S100A4 on surgical specimens suggests the need for future prospective studies of these biomarkers on biopsy specimens.
Collapse
|
32
|
Xue J, Jia E, Ren N, Lindsay A, Yu H. Circulating microRNAs as promising diagnostic biomarkers for pancreatic cancer: a systematic review. Onco Targets Ther 2019; 12:6665-6684. [PMID: 31692495 PMCID: PMC6707936 DOI: 10.2147/ott.s207963] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/16/2019] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most common forms of malignant tumors and causes of tumor-related death worldwide. The current prognosis of PC still remains poor due to the lack of effective early detection method. Recently, there is strong support that circulating miRNAs can be used as biomarkers for early detection of various cancers, including PC. The purpose of this review is to provide an overview of previous published studies on circulating miRNAs in plasma/serum for early detection of PC and summarize their diagnostic value. PubMed, Embase and Web of Science were systematically searched for eligible studies on circulating miRNAs for PC detection. Overall, 29 studies published between 2009 and 2018 evaluating 51 individual miRNAs (no P-value exceeding 0.05) and 13 miRNAs panels were included. Generally, the diagnostic performance of circulating miRNAs for PC detection was strong, with both the sensitivity and specificity of 36% individual miRNAs and 40% miRNAs panels exceeding 80%. Moreover, two promising miRNA panels were discovered and verified externally with all AUC values exceeding 0.95. Therefore, circulating miRNAs may hold potential to be used as noninvasive diagnostic biomarkers for PC, but large-scale studies are still needed to validate the promising miRNAs and optimize the miRNA panels. Since, the tremendous heterogeneity of studies in this field hampers translating miRNA markers into clinical practice, miRNA analytical procedures are also needed to be standardized in the future.
Collapse
Affiliation(s)
- Jinru Xue
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Erna Jia
- Department of Gastroenterology, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Na Ren
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Andrew Lindsay
- Major Cancer Biology, German Cancer Research Center, Heidelberg, Germany.,Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Haixin Yu
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China.,Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
33
|
Guo J, Yang WL, Pak D, Celestino J, Lu KH, Ning J, Lokshin AE, Cheng Z, Lu Z, Bast RC. Osteopontin, Macrophage Migration Inhibitory Factor and Anti-Interleukin-8 Autoantibodies Complement CA125 for Detection of Early Stage Ovarian Cancer. Cancers (Basel) 2019; 11:cancers11050596. [PMID: 31035430 PMCID: PMC6562667 DOI: 10.3390/cancers11050596] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/18/2019] [Accepted: 04/24/2019] [Indexed: 12/11/2022] Open
Abstract
Early detection of ovarian cancer promises to reduce mortality. While serum CA125 can detect more than 60% of patients with early stage (I–II) disease, greater sensitivity might be observed with a panel of biomarkers. Ten protein antigens and 12 autoantibody biomarkers were measured in sera from 76 patients with early stage (I–II), 44 patients with late stage (III–IV) ovarian cancer and 200 healthy participants in the normal risk ovarian cancer screening study. A four-biomarker panel (CA125, osteopontin (OPN), macrophage inhibitory factor (MIF), and anti-IL-8 autoantibodies) detected 82% of early stage cancers compared to 65% with CA125 alone. In early stage subjects the area under the receiver operating characteristic curve (AUC) for the panel (0.985) was significantly greater (p < 0.001) than the AUC for CA125 alone (0.885). Assaying an independent validation set of sera from 71 early stage ovarian cancer patients, 45 late stage patients and 131 healthy women, AUC in early stage disease was improved from 0.947 with CA125 alone to 0.974 with the four-biomarker panel (p = 0.015). Consequently, OPN, MIF and IL-8 autoantibodies can be used in combination with CA125 to distinguish ovarian cancer patients from healthy controls with high sensitivity. Osteopontin appears to be a robust biomarker that deserves further evaluation in combination with CA125.
Collapse
Affiliation(s)
- Jing Guo
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
- Department of Experimental Therapeutics, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Wei-Lei Yang
- Department of Experimental Therapeutics, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Daewoo Pak
- Department of Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Joseph Celestino
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Karen H Lu
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Jing Ning
- Department of Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Anna E Lokshin
- Department of Epidemiology, Pathology, Medicine, and Obstetrics/Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | - Zhongping Cheng
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Zhen Lu
- Department of Experimental Therapeutics, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Robert C Bast
- Department of Experimental Therapeutics, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
34
|
Jiang X, Hou D, Wei Z, Zheng S, Zhang Y, Li J. Extracellular and intracellular microRNAs in pancreatic cancer: from early diagnosis to reducing chemoresistance. ACTA ACUST UNITED AC 2019. [DOI: 10.1186/s41544-019-0014-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
35
|
Zubair H, Azim S, Khan MA, Patel GK, Ahmad A, Pai S, Singh S, Singh AP. Epigenetic Control of Pancreatic Carcinogenesis and Its Regulation by Natural Products. EPIGENETICS OF CANCER PREVENTION 2019:251-270. [DOI: 10.1016/b978-0-12-812494-9.00012-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
36
|
Song J, Sokoll LJ, Pasay JJ, Rubin AL, Li H, Bach DM, Chan DW, Zhang Z. Identification of Serum Biomarker Panels for the Early Detection of Pancreatic Cancer. Cancer Epidemiol Biomarkers Prev 2018; 28:174-182. [PMID: 30333219 DOI: 10.1158/1055-9965.epi-18-0483] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/31/2018] [Accepted: 10/02/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Pancreatic cancer is a deadly disease for which available biomarkers, such as CA19-9, lack the desired sensitivity and specificity for early detection. Additional biomarkers are needed to improve both its sensitivity and specificity. METHODS Multiplex immunoassays were developed for selected biomarkers using a Bio-Plex 200 system, and analytical performance was optimized. All proteins were analyzed in sera of patients diagnosed with pancreatic ductal adenocarcinoma (PDAC; n = 188) or benign pancreatic conditions (131) and healthy controls (89). The clinical performance of these markers was evaluated individually or in combination for their ability to complement CA19-9 for the early detection of pancreatic cancer. RESULTS A 6-plex immunoassay was developed with negligible cross-reactivity, wide dynamic range, recovery of 89% to 104%, and intra-assay and interassay precision of 10.2% to 19.6% and 13.7% to 29.3%, respectively. Individually, the best biomarkers to separate PDAC early stage from chronic pancreatitis or intraductal papillary mucinous neoplasm (IPMN) were CA19-9 and MIA or CA19-9 and MIC-1. Logistic regression modeling selected the two-marker panels that significantly improved the individual biomarker performance in discriminating PDAC early stage from chronic pancreatitis (AUCCA19-9+MIA = 0.86 vs. AUCCA19-9 = 0.81 or AUCMIA = 0.75 only, P < 0.05) or IPMN (AUCCA19-9+MIC-1 = 0.81 vs. AUCCA19-9 = 0.75 or AUCMIC-1 = 0.73 only, P < 0.05). It was observed that osteopontin (OPN) outperformed CA19-9 in separating IPMN from chronic pancreatitis (AUCOPN = 0.80 vs. AUCCA19-9 = 0.70, P < 0.01). CONCLUSIONS The biomarker panels evaluated by assays with high analytical performance demonstrated potential complementary values to CA19-9, warranting additional clinical validation to determine their role in early detection of pancreatic cancer. IMPACT The validated biomarker panels could lead to earlier intervention and better outcomes.
Collapse
Affiliation(s)
- Jin Song
- Department of Pathology, Center for Biomarker Discovery and Translation, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Lori J Sokoll
- Department of Pathology, Center for Biomarker Discovery and Translation, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jered J Pasay
- Department of Pathology, Center for Biomarker Discovery and Translation, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Abigail L Rubin
- Department of Pathology, Center for Biomarker Discovery and Translation, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hanying Li
- Department of Pathology, Center for Biomarker Discovery and Translation, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dylan M Bach
- Department of Pathology, School of Medicine, University of California, Irvine, California
| | - Daniel W Chan
- Department of Pathology, Center for Biomarker Discovery and Translation, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zhen Zhang
- Department of Pathology, Center for Biomarker Discovery and Translation, Johns Hopkins University School of Medicine, Baltimore, Maryland. .,Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
37
|
The ambiguous role of microRNA-205 and its clinical potential in pancreatic ductal adenocarcinoma. J Cancer Res Clin Oncol 2018; 144:2419-2431. [DOI: 10.1007/s00432-018-2755-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/18/2018] [Indexed: 12/18/2022]
|
38
|
Clinical Impact of Epithelial-to-Mesenchymal Transition Regulating MicroRNAs in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2018; 10:cancers10090328. [PMID: 30217058 PMCID: PMC6162771 DOI: 10.3390/cancers10090328] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 08/30/2018] [Accepted: 09/11/2018] [Indexed: 12/20/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive carcinoma entities worldwide with early and rapid dissemination. Recently, we discussed the role of microRNAs as epigenetic regulators of Epithelial-to-Mesenchymal Transition (EMT) in PDAC. In this study, we investigated their value as diagnostic and prognostic markers in tissue and blood samples of 185 patients including PDAC, non-malignant pancreatic disorders, and age-matched healthy controls. Expression of the microRNA-200-family (microRNAs -141, -200a, -200b, -200c, -429) and microRNA-148a was significantly downregulated in tissue of PDAC Union internationale contre le cancer (UICC) Stage II. Correspondingly, stromal PDAC tissue showed strong expression of Fibronectin, Vimentin, and ZEB-1 (Zinc finger E-box-binding homeobox) versus low expression of E-cadherin. Transient transfection of microRNA-200b and microRNA-200c mimics resulted in the downregulation of their key target ZEB-1. Inversely, blood serum analyses of patients with PDAC UICC Stages II, III, and IV showed a significant over-expression of microRNA-200-family members, microRNA-148a, microRNA-10b, and microRNA-34a. Correspondingly, Enzyme-linked Immunosorbent Assay (ELISA) analyses revealed a significant over-expression of soluble E-cadherin in serum samples of PDAC patients versus healthy controls. The best diagnostic accuracy to distinguish between PDAC and non-PDAC in this patient collective could be achieved in tissue by microRNA-148a with an area under the receiver-operating-characteristic (ROC) curve (AUC) of 0.885 and in blood serum by a panel of microRNA-141, -200b, -200c, and CA.19-9 with an AUC of 0.890. Both diagnostic tools outreach the diagnostic performance of the currently most common diagnostic biomarker CA.19-9 (AUC of 0.834). Kaplan Meier survival analysis of this patient collective revealed an improved overall survival in PDAC patients with high expression of tissue-related microRNA-34a, -141, -200b, -200c, and -429. In conclusion, EMT-regulating microRNAs have great potential as liquid and solid biopsy markers in PDAC patients. Their prognostic and therapeutic benefits remain important tasks for future studies.
Collapse
|
39
|
Giulietti M, Righetti A, Principato G, Piva F. LncRNA co-expression network analysis reveals novel biomarkers for pancreatic cancer. Carcinogenesis 2018; 39:1016-1025. [PMID: 29796634 DOI: 10.1093/carcin/bgy069] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/30/2018] [Accepted: 05/21/2018] [Indexed: 12/19/2022] Open
Abstract
High mortality and low survival rates for pancreatic ductal adenocarcinoma (PDAC) mainly result from the delay in diagnosis and treatment. Therefore, there is an urgent need to identify early PDAC biomarkers and new therapeutic targets. In this study, we applied a commonly used systems biology approach, the weighted gene co-expression network analysis (WGCNA), on lncRNA expression data. Eleven lncRNAs, namely A2M-AS1, DLEU2, LINC01133, LINC00675, MIR155HG, SLC25A25-AS1, LINC01857, LOC642852 (LINC00205), ITGB2-AS1, TSPOAP1-AS1 and PSMB8-AS1 have been identified and validated on an independent PDAC expression dataset. Furthermore, we characterized them by functional and pathway enrichment analysis and identified which lncRNAs showed differential expression, differential promoter methylation levels and copy number alterations between normal and PDAC samples. Finally, we also performed a survival analysis and identified A2M-AS1, LINC01133, LINC00205 and TSPOAP1-AS1 as prognostic biomarkers for PDAC. Interestingly, although only a few cancer-associated lncRNAs have been functionally characterized, LINC00675 and LINC01133 lncRNAs have already been demonstrated to be involved in PDAC development and progression. Therefore, our results provide new potential diagnostic/prognostic biomarkers and therapeutic targets for PDAC that deserve to be further investigated. Moreover, these lncRNAs may improve the understanding about molecular pathogenesis of PDAC.
Collapse
Affiliation(s)
- Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Alessandra Righetti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Giovanni Principato
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
40
|
Abstract
Pancreatic cancers with poor prognosis are highly malignant, readily metastatic and of immune tolerance, mainly due to delayed detection. The metastatic progression and immune tolerance of pancreatic cancer is greatly attributed to the intercellular communication. However, exosomes are deemed to be the most important tool of intercellular communicators. Thus, we present a review of pancreatic cancer and exosomes in this article. We intensively summarize the progress of early pancreatic cancer and the relationship of the proliferation, progression and metastasis of pancreatic cancer and pancreatic cancer-derived exosomes, and propose new ideas of the study of pancreatic cancer.
Collapse
Affiliation(s)
- Chengfei Zhao
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350108, Fujian, China.,Department of Pharmacy, Pharmacy and Medical Technology School, Putian University, Putian 351100, Fujian, China.,Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350108, Fujian, China
| | - Feng Gao
- Department of Pathology, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China.,Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350108, Fujian, China
| | - Shaohuang Weng
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350108, Fujian, China
| | - Qicai Liu
- Department of Clinical Laboratory, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| |
Collapse
|
41
|
Systematic review on the role of serum tumor markers in the detection of recurrent pancreatic cancer. HPB (Oxford) 2018; 20:297-304. [PMID: 29366815 DOI: 10.1016/j.hpb.2017.11.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/22/2017] [Accepted: 11/26/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Biomarker testing can be helpful to monitor disease progression after resection of pancreatic cancer. This systematic review aims to give an overview of the literature on the diagnostic value of serum tumor markers for the detection of recurrent pancreatic cancer during follow-up. METHODS A systematic search was performed to 2 October 2017. All studies reporting on the diagnostic value of postoperatively measured serum biomarkers for the detection of pancreatic cancer recurrence were included. Data on diagnostic accuracy of tumor markers were extracted. Forest plots and pooled values of sensitivity and specificity were calculated. RESULTS Four articles described test results of CA 19-9. A pooled sensitivity and specificity of respectively 0.73 (95% CI 0.66-0.80) and 0.83 (95% CI 0.73-0.91) were calculated. One article reported on CEA, showing a sensitivity of 50% and specificity of 65%. No other serum tumor markers were discussed for surveillance purposes in the current literature. CONCLUSION Although testing of serum CA 19-9 has considerable limitations, CA 19-9 remains the most used serum tumor marker for surveillance after surgical resection of pancreatic cancer. Further studies are needed to assess the role of serum tumor marker testing in the detection of recurrent pancreatic cancer and to optimize surveillance strategies.
Collapse
|
42
|
Armstrong EA, Beal EW, Chakedis J, Paredes AZ, Moris D, Pawlik TM, Schmidt CR, Dillhoff ME. Exosomes in Pancreatic Cancer: from Early Detection to Treatment. J Gastrointest Surg 2018; 22:737-750. [PMID: 29423813 DOI: 10.1007/s11605-018-3693-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/12/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Pancreatic cancer (PC) remains one of the most fatal forms of cancer worldwide with incidence nearly equal to mortality. This is often attributed to the fact that diagnosis is often not made until later disease stages when treatment proves difficult. Efforts have been made to reduce the mortality of PC through improvements in early screening techniques and treatments of late-stage disease. Exosomes, small extracellular vesicles involved in cellular communication, have shown promise in helping understand PC disease biology. METHODS In this review, we discuss current studies of the role of exosomes in PC physiology, and their potential use as diagnostic and treatment tools. RESULTS Exosomes have a role in diagnosing pancreatic cancer and in understanding tumor biology including migration, proliferation, chemoresistance, immunosuppression, cachexia and diabetes, and have a potential role in therapy for pancreatic cancer. CONCLUSIONS Exosomal analysis is beneficial in demonstrating mechanisms behind PC growth and metastasis, immunosuppression, drug resistance, and paraneoplastic conditions. Furthermore, the use of exosomes can be beneficial in detecting early-stage PC and exosomes have potential applications as therapeutic targets.
Collapse
Affiliation(s)
- Emily A Armstrong
- The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Eliza W Beal
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA.
| | - Jeffery Chakedis
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Anghela Z Paredes
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Demetrios Moris
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Carl R Schmidt
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Mary E Dillhoff
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| |
Collapse
|
43
|
Dumstrei K, Chen H, Brenner H. A systematic review of serum autoantibodies as biomarkers for pancreatic cancer detection. Oncotarget 2017; 7:11151-64. [PMID: 26840568 PMCID: PMC4905464 DOI: 10.18632/oncotarget.7098] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/18/2016] [Indexed: 12/22/2022] Open
Abstract
Pancreatic cancer is a leading cause of cancer-related deaths in the western world. Patients with pancreatic cancer have poor prognosis, partly due to difficulties in detecting it at early stages. While different markers have been associated with pancreatic cancer, many of them show suboptimal sensitivity and specificity. Serum autoantibodies against tumor-associated antigens have recently emerged as early stage biomarkers for different types of cancers. Given the urgent need for early and reliable biomarkers for pancreatic cancer, we undertook a systematic review of the published literature to identify primary articles that evaluated serum autoantibodies in pancreatic cancer detection by searching PubMed and ISI Web of Knowledge. Two reviewers extracted data on study characteristics and results independently. Overall, 31 studies evaluating 124 individual serum autoantibodies in pancreatic cancer detection met the inclusion criteria. In general, single autoantibody markers showed relatively low sensitivities at high specificity. A combination of markers, either multiple serum autoantibodies or serum autoantibodies combined with tumor-associated markers, led to a better diagnostic performance. However, most of the analyzed autoantibodies have only been reported in single studies and therefore need to be independently validated. We conclude that serum autoantibodies might present an option as biomarkers for early detection of pancreatic cancer, but more work is needed to identify and validate autoantibody signatures that are associated with early stage pancreatic cancer.
Collapse
Affiliation(s)
- Karin Dumstrei
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany.,European Molecular Biology Organization (EMBO), D-69117 Heidelberg, Germany
| | - Hongda Chen
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany.,Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), D-69120 Heidelberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| |
Collapse
|
44
|
Preoperative Carbohydrate Antigen 19-9: Its Neglected Role in Alpha-Fetoprotein-Negative Hepatocellular Carcinoma Patients. J Gastrointest Surg 2017; 21:2025-2032. [PMID: 28840443 DOI: 10.1007/s11605-017-3528-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/30/2017] [Indexed: 01/31/2023]
Abstract
PURPOSE The study aimed to investigate the prognostic role of the preoperative carbohydrate antigen 19-9 (CA19-9) level in alpha-fetoprotein (AFP)-negative (AFP < 25 ng/ml) hepatocellular carcinoma (HCC) patients. METHODS From December 2004 to December 2013, 750 patients diagnosed with AFP-negative HCC following curative resection were enrolled. Both univariate and multivariate analyses were performed to clarify the prognostic factors for overall survival (OS) and disease-free survival (DFS). RESULTS The optimal cutoff point for preoperative CA19-9 was 32.6 U/ml, and the value of the area under the curve was 0.640. The 1-, 3-, and 5-year OS rates were 88.4, 72.2, and 57.0%, respectively, for the CA19-9 > 32.6 U/ml group and 97.0, 83.3, and 79.9%, respectively, for the CA19-9 ≤ 32.6 U/ml group (P < 0.001). The 1-, 3-, and 5-year DFS rates were 71.8, 47.7, and 34.8%, respectively, for the CA19-9 > 32.6 U/ml group and 80.8, 63.6, and 55.5%, respectively, for the CA19-9 ≤ 32.6 U/ml group (P < 0.001). The multivariate analysis showed that CA19-9 > 32.6 U/ml was one of the most significant unfavorable predictors of OS and DFS (P < 0.001 for both). CONCLUSIONS Preoperative CA19-9 > 32.6 U/ml is a predictor of dismal prognosis and can be employed as a prognostic marker for patient selection in AFP-negative HCC management.
Collapse
|
45
|
Schiergens TS, Renz BW, Reu S, Neumann J, Al-Sayegh R, Nieß H, Ilmer M, Kruger S, Boeck S, Heinemann V, Werner J, Kleespies A. Prognostic Value of Preoperative Serum Carcinoembryonic Antigen and Carbohydrate Antigen 19-9 After Resection of Ampullary Cancer. J Gastrointest Surg 2017; 21:1775-1783. [PMID: 28875420 DOI: 10.1007/s11605-017-3489-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/27/2017] [Indexed: 01/31/2023]
Abstract
BACKGROUND The purpose of this study is to investigate the prognostic value of pre-resection serum carcinoembryonic antigen (CEA) and carbohydrate antigen (CA) 19-9 after resection of ampullary cancer (AC) in consideration of intestinal (IT) and pancreatobiliary (PT) subtypes. METHODS Overall survival (OS) analysis of patients undergoing curative resection of ampullary cancer. RESULTS Elevated preoperative CEA (P = 0.013) and CA 19-9 levels (P = 0.030) were significant prognostic factors. Subgroup analysis, however, showed both markers having prognostic value only for the IT subgroup. Pre-resection CEA within normal range identified a subgroup of IT patients with an excellent median survival of 145 months. Compared to other AC patients, this low-risk ITCEA- subpopulation was characterized by less frequent advanced pT stages (pT3/pT4, 41 vs. 62%; P = 0.047) and lymph node involvement (pN+, 30 vs. 65%; P = 0.001). OS of this subgroup was significantly better compared to other AC patients (145 vs. 25 months; HR = 3.8; P < 0.001). By multivariate survival analysis, the patient age, the PT subtype, and an elevated pre-resection serum CEA value were identified as independent prognostic variables. CONCLUSIONS In AC, the histomorphologic subclassification is highly relevant regarding the prognostic value of preoperative serum CEA and CA 19-9. IT-patients with normal preoperative CEA represent a favorable subgroup with excellent long-term survival.
Collapse
Affiliation(s)
- Tobias S Schiergens
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University of Munich, Campus Grosshadern, Munich, Germany
| | - Bernhard W Renz
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University of Munich, Campus Grosshadern, Munich, Germany
| | - Simone Reu
- Department of Pathology, University of Munich, Munich, Germany
| | - Jens Neumann
- Department of Pathology, University of Munich, Munich, Germany
| | - Rami Al-Sayegh
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University of Munich, Campus Grosshadern, Munich, Germany
| | - Hanno Nieß
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University of Munich, Campus Grosshadern, Munich, Germany
| | - Matthias Ilmer
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University of Munich, Campus Grosshadern, Munich, Germany
| | - Stephan Kruger
- Department of Medical Oncology, University of Munich, Campus Grosshadern, Munich, Germany
| | - Stefan Boeck
- Department of Medical Oncology, University of Munich, Campus Grosshadern, Munich, Germany
| | - Volker Heinemann
- Department of Medical Oncology, University of Munich, Campus Grosshadern, Munich, Germany
| | - Jens Werner
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University of Munich, Campus Grosshadern, Munich, Germany
| | - Axel Kleespies
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University of Munich, Campus Grosshadern, Munich, Germany.
| |
Collapse
|
46
|
Burgener JM, Rostami A, De Carvalho DD, Bratman SV. Cell-free DNA as a post-treatment surveillance strategy: current status. Semin Oncol 2017; 44:330-346. [DOI: 10.1053/j.seminoncol.2018.01.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 01/18/2018] [Accepted: 01/31/2018] [Indexed: 02/06/2023]
|
47
|
Mustafa S, Pan L, Marzoq A, Fawaz M, Sander L, Rückert F, Schrenk A, Hartl C, Uhler R, Yildirim A, Strobel O, Hackert T, Giese N, Büchler MW, Hoheisel JD, Alhamdani MSS. Comparison of the tumor cell secretome and patient sera for an accurate serum-based diagnosis of pancreatic ductal adenocarcinoma. Oncotarget 2017; 8:11963-11976. [PMID: 28060763 PMCID: PMC5355318 DOI: 10.18632/oncotarget.14449] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 12/25/2016] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer is the currently most lethal malignancy. Toward an accurate diagnosis of the disease in body liquids, we studied the protein composition of the secretomes of 16 primary and established cell lines of pancreatic ductal adenocarcinoma (PDAC). Compared to the secretome of non-tumorous cells, 112 proteins exhibited significantly different abundances. Functionally, the proteins were associated with PDAC features, such as decreased apoptosis, better cell survival and immune cell regulation. The result was compared to profiles obtained from 164 serum samples from two independent cohorts - a training and a test set - of patients with PDAC or chronic pancreatitis and healthy donors. Eight of the 112 secretome proteins exhibited similar variations in their abundance in the serum profile specific for PDAC patients, which was composed of altogether 189 proteins. The 8 markers shared by secretome and serum yielded a 95.1% accuracy of distinguishing PDAC from healthy in a Receiver Operating Characteristic curve analysis, while any number of serum-only markers produced substantially less accurate results. Utility of the identified markers was confirmed by classical enzyme linked immunosorbent assays (ELISAs). The study highlights the value of cell secretome analysis as a means of defining reliable serum biomarkers.
Collapse
Affiliation(s)
- Shakhawan Mustafa
- Division of Functional Genome Analysis, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany.,Kurdistan Institution for Strategic Studies and Scientific Research, Kurdistan Region, Iraq
| | - Longqiang Pan
- Division of Functional Genome Analysis, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Aseel Marzoq
- Division of Functional Genome Analysis, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Malak Fawaz
- Division of Functional Genome Analysis, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Laureen Sander
- Division of Functional Genome Analysis, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Felix Rückert
- Chirurgische Klinik, Universitätsmedizin Mannheim, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany
| | - Andrea Schrenk
- Chirurgische Klinik, Universitätsmedizin Mannheim, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany
| | - Christina Hartl
- Division of Functional Genome Analysis, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Rico Uhler
- Division of Functional Genome Analysis, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Adem Yildirim
- Division of Functional Genome Analysis, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Oliver Strobel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Nathalia Giese
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Markus W Büchler
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jörg D Hoheisel
- Division of Functional Genome Analysis, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | | |
Collapse
|
48
|
Chen D, Wu X, Xia M, Wu F, Ding J, Jiao Y, Zhan Q, An F. Upregulated exosomic miR‑23b‑3p plays regulatory roles in the progression of pancreatic cancer. Oncol Rep 2017; 38:2182-2188. [PMID: 28849236 PMCID: PMC5652966 DOI: 10.3892/or.2017.5919] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/14/2017] [Indexed: 12/21/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most lethal malignances. Identification of biomarkers for early diagnosis of PC is a key imperative. MicroRNAs (miRNAs) have been shown to be valuable biomarkers in the context of several cancers. Exosomes refer to vesicles released by the tumor cells at the early stage of disease. Thus, detection of miRNA in exosomes can be used as a potential biomarker for PC. In this study, we profiled serum levels of miRNAs in patients with chronic pancreatitis (CP) and PC; the role of miR-23b-3p in PC progression was assessed in vitro. Additionally, we assessed, the expression of miR-23b-3p in exosomes isolated from serum samples and assessed the correlation between the expression of miR-23b-3p and carbohydrate antigen 19-9 (CA19-9). Three serum samples each were randomly selected from healthy controls (n=20), and patients with CP (n=18) and PC (n=16) for miRNA microarray profiling. The dysregulated miRNAs were confirmed using qRT-PCR. Four dysregulated miRNAs common to patients with CP and PC were identified on miRNA microarray analysis and confirmed by qRT-PCR. miR-23b-3p level was consistently higher in serum samples from PC patients as compared to those from healthy controls and CP patients (p<0.05). Overexpression of miR-23b-3p promoted proliferation, migration, and invasion capability of PC cells in vitro (p<0.05). Furthermore, miR-23b-3p was upregulated in exosomes of PC serum samples and the supernatant of pancreatic cancer cells (PANC-1), and the expression levels of miR-23b-3p were associated with those of serum CA19-9 levels. This study provides insights into the potential role of miR-23b-3p as a novel biomarker and target for treatment of PC.
Collapse
Affiliation(s)
- Dayang Chen
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Xiongbo Wu
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Min Xia
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Fang Wu
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Junli Ding
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Yang Jiao
- School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Medical School of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Qiang Zhan
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Fangmei An
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| |
Collapse
|
49
|
Drug-eluting scaffold inhibited in vivo pancreatic tumorigenesis by engaging murine CCR4 +CD8 + T cells. Colloids Surf B Biointerfaces 2017; 158:469-473. [PMID: 28732312 DOI: 10.1016/j.colsurfb.2017.07.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 07/02/2017] [Accepted: 07/07/2017] [Indexed: 01/08/2023]
Abstract
CCL17 is well known for its ability to engage CCR4+CD8+ T cells, which have been shown to play a critical role in preventing tumorigenesis. In this study, we attempted to inhibit in vivo pancreatic tumorigenesis by engaging murine CCR4+CD8+ T cells through a drug-eluting scaffold with a payload of CCL17. The drug-eluting scaffold was fabricated by electrospinning polyglyconate and porcine gelatin. The electrospun scaffold featured randomly distributed non-woven fibers with diameters ranging from 1μm to 4μm. The in vitro study confirmed that scaffolding materials were non-cytotoxic to pancreatic cancer cells. The in vivo study showed an increased presence of murine CCR4+CD8+ T cells into the tumor mass treated with drug-eluting scaffold compared to those with non-eluting scaffold or the control groups. The weights of tumor masses were 132.04mg±12.25mg in the control group, 158.12mg±18.98mg in the NES group and 96.22mg±14.56mg in the DES group, respectively. The volumes of tumor masses were 1035.21mm3±128.97mm3 in the control group, 978.56mm3±110.19mm3 in the NES group and 634.35mm3±87.12mm3 in the DES group, respectively. Further study showed that the increased presence of CCR4+CD8+ T cells also inhibited the hepatic metastasis of pancreatic cancer cells. Our study shed a new light on the post-operative treatment of pancreatic cancer to prevent the recurrence.
Collapse
|
50
|
Erb U, Zhao K, Wang Z, Xiao L, Zöller M. Murine and human pancreatic tumor exosome recovery in mouse serum: Diagnostic and prognostic potential and target cell delivery. Cancer Lett 2017; 403:1-12. [PMID: 28619525 DOI: 10.1016/j.canlet.2017.06.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/26/2017] [Accepted: 06/03/2017] [Indexed: 12/12/2022]
Abstract
Exosomes (Exo), powerful intercellular communicators, are recovered in all body fluids, suggesting suitability for diagnosis and prognosis. Easy in vitro manipulation recommends Exo as drug vehicles. Aiming to consolidate diagnostic and therapeutic potential of Exo, we evaluated recovery and fate of tumor (TEX) and exogenous Exo in syngeneic and xenogeneic mice bearing a murine or a human pancreatic adenocarcinoma. A significant increase in serum (S)-TEX was observed 2 weeks after tumor cell application. Instead, S-TEX declined within 3-6 days after tumor excision. Intravenously injected dye-labeled TEX were rapidly cleared from the serum. Partly being degraded in the liver, the majority is taken-up by PBL, liver, bone marrow and lung cells. In the tumor-bearing host TEX persisted longer becoming enriched in tumor cells and metastatic organs. Accordingly, an antibody blockade of a TEX marker hampered disseminated tumor cell settlement in selected organs. In brief, a tumor marker panel appears suited for S-TEX recovery. In murine models, S-TEX are qualified for therapy control and follow-up studies. Despite rapid clearance from the serum, Exo uptake by host cells is most promising for tailored Exo as drug transporter.
Collapse
Affiliation(s)
- Ulrike Erb
- Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | - Kun Zhao
- Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | - Zhe Wang
- Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | - Li Xiao
- Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | - Margot Zöller
- Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany.
| |
Collapse
|