1
|
Lv TR, Wang JM, Ma WJ, Hu YF, Dai YS, Jin YW, Li FY. The consistencies and inconsistencies between distal cholangiocarcinoma and pancreatic ductal adenocarcinoma: A systematic review and meta-analysis. Front Oncol 2022; 12:1042493. [PMID: 36578941 PMCID: PMC9791204 DOI: 10.3389/fonc.2022.1042493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/09/2022] [Indexed: 12/14/2022] Open
Abstract
Objective To evaluate the consistencies and inconsistencies between distal cholangiocarcinoma (DCCA) and pancreatic ductal adenocarcinoma (PDCA) regarding their biological features and long-term prognosis. Methods PubMed, the Cochrane Library, and EMBASE were searched to find comparative studies between DCCA and PDCA. RevMan5.3 and Stata 13.0 software were used for the statistical analyses. Results Eleven studies with 4,698 patients with DCCA and 100,629 patients with PDCA were identified. Pooled results indicated that patients with DCCA had a significantly higher rate of preoperative jaundice (p = 0.0003). Lymphatic metastasis (p < 0.00001), vascular invasion (p < 0.0001), and peri-neural invasion (p = 0.005) were more frequently detected in patients with PDCA. After curative pancreaticoduodenectomy (PD), a significantly higher R0 rate (p < 0.0001) and significantly smaller tumor size (p < 0.00001) were detected in patients with DCCA. Patients with DCCA had a more favorable overall survival (OS) (p < 0.00001) and disease-free survival (DFS) (p = 0.005) than patients with PDCA. However, postoperative morbidities (p = 0.02), especially postoperative pancreatic fistula (POPF) (p < 0.00001), more frequently occurred in DCCA. Conclusion Patients with DCCA had more favorable tumor pathological features and long-term prognosis than patients with PDCA. An early diagnosis more frequently occurred in patients with DCCA. However, postoperative complications, especially POPF, were more frequently observed in patients with DCCA.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fu-Yu Li
- *Correspondence: Yan-Wen Jin, ; Fu-Yu Li,
| |
Collapse
|
2
|
Pancreatic Cancer Cells Induce MicroRNA Deregulation in Platelets. Int J Mol Sci 2022; 23:ijms231911438. [PMID: 36232741 PMCID: PMC9569638 DOI: 10.3390/ijms231911438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer is a pathology with a high mortality rate since it is detected at advanced stages, so the search for early-stage diagnostic biomarkers is essential. Liquid biopsies are currently being explored for this purpose and educated platelets are a good candidate, since they are known to present a bidirectional interaction with tumor cells. In this work, we analyzed the effects of platelets on cancer cells’ viability, as determined by MTT, migration using transwell assays, clonogenicity in soft agar and stemness by dilution assays and stem markers’ expression. We found that the co-culture of platelets and pancreatic cancer cells increased the proliferation and migration capacity of BXCP3 cells, augmented clonogenicity and induced higher levels of Nanog, Sox2 and Oct4 expression. As platelets can provide horizontal transfer of microRNAs, we also determined the differential expression of miRNAs in platelets obtained from a small cohort of pancreatic cancer patients and healthy subjects. We found clear differences in the expression of several miRNAs between platelets of patients with cancer healthy subjects. Moreover, when we analyzed microRNAs from the platelets of the pancreatic juice and blood derived from each of the cancer patients, interestingly we find differences between the blood- and pancreatic juice-derived platelets suggesting the presence of different subpopulations of platelets in cancer patients, which warrant further analysis.
Collapse
|
3
|
Mori H, Tanoue S, Takaji R, Ueda S, Okahara M, Ueda SS. Arterial Administration of DNA Crosslinking Agents with Restraint of Homologous Recombination Repair by Intravenous Low-Dose Gemcitabine Is Effective for Locally Advanced Pancreatic Cancer. Cancers (Basel) 2022; 14:cancers14010220. [PMID: 35008384 PMCID: PMC8750330 DOI: 10.3390/cancers14010220] [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: 11/11/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Pancreatic cancer is considered incurable, and most cases are detected in the advanced stages. Establishing a new, effective interventional treatment for advanced pancreatic cancer is a pressing issue. Pretreatment with gemcitabine had a restraining effect on the homologous DNA recombination repair (HRR) of DNA crosslinking, inhibiting the function of Rad51, of which expression is found to be increased in pancreatic cancer. The aim of our prospective study was to assess the potential value of the arterial administration of DNA crosslinking agents after intravenous administration of low-dose gemcitabine for patients with advanced pancreatic cancer. We confirmed, among forty-five patients with unresectable advanced pancreatic cancer, that a patient subgroup of locally advanced pancreatic cancer (LAPC, 10 patients) who underwent these treatment courses successively more than twice in the first 6 months had 33 months of overall survival, 31 months of local progression free survival, and a complete response of 40%. This treatment can be a new treatment option for LAPC. Abstract (1) Background: Pretreatment by Rad51-inhibitory substances such as gemcitabine followed by arterial chemotherapy using antineoplastic agents causing DNA crosslink might be more beneficial for patients with locally advanced pancreatic cancers than conventional treatments. The efficacy of arterial administration of DNA crosslinking agents with pretreatment of intravenous low-dose gemcitabine for patients with unresectable locally advanced or metastatic pancreatic cancer (LAPC or MPC) is evaluated. (2) Methods: A single-arm, single-center, institutional review board-approved prospective study was conducted between 2005 and 2015. Forty-five patients (23 LAPC, 22 MPC) were included. Patients received a weekly low dose of gemcitabine intravenously for three weeks followed by arterial administration of mitomycin C and epirubicin hydrochloride at tumor-supplying arteries on the fifth or sixth week. This treatment course was repeated at 1.5-to-2-month intervals. Overall survival (OS), local progression-free survival (LPFS), and therapeutic response were evaluated. LAPC or MPC were divided according to treatment compliance, excellent or poor (1 or 2), to subgroups L1, L2, M1, and M2. (3) Results: OS of LAPC and MPC were 23 months and 13 months, respectively. The OS of LAPC with excellent treatment compliance (subgroup L1, 10 patients) was 33 months with 31 months of LPFS, and four patients (40%) had a complete response (CR). The OS of the L1 subgroup was significantly longer than those of other subgroups L2, M1, and M2, which were 17 months, 17 months, and 8 months, respectively. As Grade 3 adverse effects, severe bone marrow suppression, interstitial pneumonitis, and hemolytic uremic syndrome were observed in six (13.0%), three (6.5%), and three (6.5%) patients, respectively. (4) Conclusions: Arterial DNA crosslinking with the systemic restraint of homologous recombination repair can be a new treatment option for LAPC.
Collapse
Affiliation(s)
- Hiromu Mori
- Department of Radiology, Nagato Memorial Hospital, Saiki 876-0835, Japan;
- Department of Radiology, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (S.T.); (R.T.); (M.O.)
- Correspondence: ; Tel.: +81-80-4270-0753
| | - Shuichi Tanoue
- Department of Radiology, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (S.T.); (R.T.); (M.O.)
- Department of Radiology, School of Medicine, Kurume University, Kurume 830-0011, Japan
| | - Ryo Takaji
- Department of Radiology, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (S.T.); (R.T.); (M.O.)
| | - Shinya Ueda
- Department of Radiology, Nagato Memorial Hospital, Saiki 876-0835, Japan;
- San-Ai Medical Center, Department of Radiology, Oita 870-1151, Japan
- Department of Radiology, Shin-Beppu Hospital, Beppu 874-8538, Japan
| | - Mika Okahara
- Department of Radiology, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (S.T.); (R.T.); (M.O.)
- Department of Radiology, Shin-Beppu Hospital, Beppu 874-8538, Japan
| | - Saori Sugi Ueda
- Department of Gastroenterology, Shin-Beppu Hospital, Beppu 874-8538, Japan;
- San-Ai Medical Center, Department of Gastroenterology, Oita 870-115, Japan
| |
Collapse
|
4
|
Khachfe HH. Use of liquid biopsies in gastrointestinal cancers. World J Gastrointest Oncol 2021; 13:1210-1212. [PMID: 34616524 PMCID: PMC8465448 DOI: 10.4251/wjgo.v13.i9.1210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/16/2021] [Accepted: 08/09/2021] [Indexed: 02/06/2023] Open
Abstract
The use of liquid biopsies is a relatively new tool in diagnosis and management of gastrointestinal cancers and is actively being investigated. Liquid biopsies have become extremely popular in cholangiocarcinoma and pancreatic cancer research. With more prospective trials using this tool for early diagnosis, liquid biopsies may become an important part of cancer management.
Collapse
Affiliation(s)
- Hussein H Khachfe
- Department of Gastrointestinal Surgical Oncology, University of Pittsburgh, Pittsburgh, PA 15232, United States
| |
Collapse
|
5
|
Wang SE, Shyr BU, Shyr BS, Chen SC, Chang SC, Shyr YM. Circulating Cell-Free DNA in Pancreatic Head Adenocarcinoma Undergoing Pancreaticoduodenectomy. Pancreas 2021; 50:214-218. [PMID: 33565798 DOI: 10.1097/mpa.0000000000001730] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Circulating cell-free DNA (cfDNA) analysis is recently reported as a promising prognostic biomarker in various types of cancer. This study aimed to evaluate the role of cfDNA in pancreatic head adenocarcinoma. METHODS Data for pancreatic head adenocarcinoma undergoing pancreaticoduodenectomy were studied for cfDNA. Prognostic factors were determined, and their correlation with cfDNA level was evaluated. RESULTS The median of cfDNA for 97 cases was 7724 copies/mL, with a mean of 10,467, and ranging from 1856 to 44,203. Cell-free DNA was significantly higher in positive lymph node involvement and advanced stage III. Poor prognostic factors included high cfDNA level (>7724 copies/mL), abnormal carbohydrate antigen 19-9, abnormal carcinoembryonic antigen, and advanced stage. The 1- and 5-year survivals for those with high cfDNA were poorer, 70.2% and 21.2%, respectively, as compared with 93.4% and 23.7% for those with low cfDNA level. Only cfDNA level and stage were independent prognostic factors after multivariate analysis. CONCLUSIONS The level of cfDNA was correlated with tumor burden. Therefore, it could be an emerging survival predictor for resectable pancreatic head adenocarcinoma, and its detection might be a promising liquid biopsy to monitor both tumor progression and treatment response.
Collapse
Affiliation(s)
- Shin-E Wang
- From the Department of Surgery, Taipei Veterans General Hospital, National Yang Ming University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
6
|
Shah D, Lamarca A, Valle JW, McNamara MG. The Potential Role of Liquid Biopsies in Advancing the Understanding of Neuroendocrine Neoplasms. J Clin Med 2021; 10:jcm10030403. [PMID: 33494364 PMCID: PMC7865482 DOI: 10.3390/jcm10030403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 12/22/2022] Open
Abstract
Tumour tissue as a source for molecular profiling and for in vivo models has limitations (e.g., difficult access, limited availability, single time point, potential heterogeneity between primary and metastatic sites). Conversely, liquid biopsies provide an easily accessible approach, enabling timely and longitudinal interrogation of the tumour molecular makeup, with increased ability to capture spatial and temporal intra-tumour heterogeneity compared to tumour tissue. Blood-borne biomarker assays (e.g., circulating tumour cells (CTCs), circulating free/tumour DNA (cf/ctDNA)) pose unique opportunities for aiding in the molecular characterisation and phenotypic subtyping of neuroendocrine neoplasms and will be discussed in this article.
Collapse
Affiliation(s)
- Dinakshi Shah
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK; (D.S.); (A.L.)
| | - Angela Lamarca
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK; (D.S.); (A.L.)
- Division of Cancer Sciences, University of Manchester, Manchester M13 9PL, UK;
| | - Juan W Valle
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK; (D.S.); (A.L.)
- Division of Cancer Sciences, University of Manchester, Manchester M13 9PL, UK;
| | - Mairéad G McNamara
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK; (D.S.); (A.L.)
- Division of Cancer Sciences, University of Manchester, Manchester M13 9PL, UK;
- Correspondence:
| |
Collapse
|
7
|
Driescher C, Fuchs K, Haeberle L, Goering W, Frohn L, Opitz FV, Haeussinger D, Knoefel WT, Keitel V, Esposito I. Bile-Based Cell-Free DNA Analysis Is a Reliable Diagnostic Tool in Pancreatobiliary Cancer. Cancers (Basel) 2020; 13:E39. [PMID: 33375555 PMCID: PMC7818177 DOI: 10.3390/cancers13010039] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
Currently available serum biomarkers for pancreatobiliary cancers lack sensitivity and specificity and ultimate diagnosis still requires invasive procedures for histological confirmation. The detection of tumor-specific genetic aberrations with utilization of cell free DNA (cfDNA) is a less invasive approach than traditional tissue biopsies; however, it has not been implemented into clinical routine. In this study, we investigated bile as a liquid biopsy source in pancreatobiliary cancers and compared its potential as cell-free DNA source to plasma. Blood (n = 37) and bile (n = 21) samples were collected from patients affected by pancreatic ductal adenocarcinoma (PDAC) and extrahepatic cholangiocarcinoma (CCA) or with non-malignant biliary obstructions (blood n = 16; bile n = 21). Panel-based next generation sequencing (NGS) and digital droplet PCR (ddPCR) were applied for tumor mutation profiling. NGS results from matched tumor tissues (n = 29) served as comparison. Sequencing of cfDNA from bile resulted in detection of 96.2% of the pathogenic tumor mutations found in matched tissue samples. On the other hand, only 31.6% of pathogenic tumor mutations found in tissue could be detected in plasma. In a direct comparison, only half of the mutations detected in bile cfDNA were concordantly detected in plasma from the same patients. Panel NGS and ddPCR displayed comparable sensitivity. In conclusion, bile is a suitable source of cfDNA for the diagnosis of pancreatobiliary cancer and performs more reliably than plasma. Although primary diagnosis still requires histologic confirmation, bile-derived cfDNA could offer an alternative if tissue sampling is not feasible and might allow less invasive disease monitoring.
Collapse
Affiliation(s)
- Caroline Driescher
- Institute of Pathology, Heinrich-Heine-University and University Hospital of Duesseldorf, 40225 Duesseldorf, Germany; (C.D.); (L.H.); (W.G.); (L.F.); (F.V.O.)
| | - Katharina Fuchs
- Department of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University and University Hospital of Duesseldorf, 40225 Duesseldorf, Germany; (K.F.); (D.H.); (V.K.)
| | - Lena Haeberle
- Institute of Pathology, Heinrich-Heine-University and University Hospital of Duesseldorf, 40225 Duesseldorf, Germany; (C.D.); (L.H.); (W.G.); (L.F.); (F.V.O.)
| | - Wolfgang Goering
- Institute of Pathology, Heinrich-Heine-University and University Hospital of Duesseldorf, 40225 Duesseldorf, Germany; (C.D.); (L.H.); (W.G.); (L.F.); (F.V.O.)
| | - Lisa Frohn
- Institute of Pathology, Heinrich-Heine-University and University Hospital of Duesseldorf, 40225 Duesseldorf, Germany; (C.D.); (L.H.); (W.G.); (L.F.); (F.V.O.)
| | - Friederike V. Opitz
- Institute of Pathology, Heinrich-Heine-University and University Hospital of Duesseldorf, 40225 Duesseldorf, Germany; (C.D.); (L.H.); (W.G.); (L.F.); (F.V.O.)
| | - Dieter Haeussinger
- Department of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University and University Hospital of Duesseldorf, 40225 Duesseldorf, Germany; (K.F.); (D.H.); (V.K.)
| | - Wolfram Trudo Knoefel
- Department of General, Thoracic and Pediatric Surgery, Heinrich-Heine-University and University Hospital of Duesseldorf, 40225 Duesseldorf, Germany;
| | - Verena Keitel
- Department of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University and University Hospital of Duesseldorf, 40225 Duesseldorf, Germany; (K.F.); (D.H.); (V.K.)
| | - Irene Esposito
- Institute of Pathology, Heinrich-Heine-University and University Hospital of Duesseldorf, 40225 Duesseldorf, Germany; (C.D.); (L.H.); (W.G.); (L.F.); (F.V.O.)
| |
Collapse
|
8
|
Iovanna J. Implementing biological markers as a tool to guide clinical care of patients with pancreatic cancer. Transl Oncol 2020; 14:100965. [PMID: 33248412 PMCID: PMC7704461 DOI: 10.1016/j.tranon.2020.100965] [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: 10/12/2020] [Revised: 11/04/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
A major obstacle for the effective treatment of PDAC is its molecular heterogeneity. Stratification of PDAC using markers highly specific, reproducible, sensitive, easily measurable and inexpensive is necessary. At the early stages, clinician’s priority lies in rapid diagnosis, so that the patient receives surgery without delay. At advanced disease stages, priority is to determine the tumor subtype and select a suitable effective treatment.
A major obstacle for the effective treatment of pancreatic ductal adenocarcinoma (PDAC) is its molecular heterogeneity, reflected by the diverse clinical outcomes and responses to therapies that occur. The tumors of patients with PDAC must therefore be closely examined and classified before treatment initiation in order to predict the natural evolution of the disease and the response to therapy. To stratify patients, it is absolutely necessary to identify biological markers that are highly specific and reproducible, and easily measurable by inexpensive sensitive techniques. Several promising strategies to find biomarkers are already available or under development, such as the use of liquid biopsies to detect circulating tumor cells, circulating free DNA, methylated DNA, circulating RNA, and exosomes and extracellular vesicles, as well as immunological markers and molecular markers. Such biomarkers are capable of classifying patients with PDAC and predicting their therapeutic sensitivity. Interestingly, developing chemograms using primary cell lines or organoids and analyzing the resulting high-throughput data via artificial intelligence would be highly beneficial to patients. How can exploiting these biomarkers benefit patients with resectable, borderline resectable, locally advanced, and metastatic PDAC? In fact, the utility of these biomarkers depends on the patient's clinical situation. At the early stages of the disease, the clinician's priority lies in rapid diagnosis, so that the patient receives surgery without delay; at advanced disease stages, where therapeutic possibilities are severely limited, the priority is to determine the PDAC tumor subtype so as to estimate the clinical outcome and select a suitable effective treatment.
Collapse
Affiliation(s)
- Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, France.
| |
Collapse
|
9
|
Ciernikova S, Earl J, García Bermejo ML, Stevurkova V, Carrato A, Smolkova B. Epigenetic Landscape in Pancreatic Ductal Adenocarcinoma: On the Way to Overcoming Drug Resistance? Int J Mol Sci 2020; 21:ijms21114091. [PMID: 32521716 PMCID: PMC7311973 DOI: 10.3390/ijms21114091] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/02/2020] [Accepted: 06/06/2020] [Indexed: 12/24/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive solid malignancies due to the rapid rate of metastasis and high resistance to currently applied cancer therapies. The complex mechanism underlying the development and progression of PDAC includes interactions between genomic, epigenomic, and signaling pathway alterations. In this review, we summarize the current research findings on the deregulation of epigenetic mechanisms in PDAC and the influence of the epigenome on the dynamics of the gene expression changes underlying epithelial–mesenchymal transition (EMT), which is responsible for the invasive phenotype of cancer cells and, therefore, their metastatic potential. More importantly, we provide an overview of the studies that uncover potentially actionable pathways. These studies provide a scientific basis to test epigenetic drug efficacy in synergy with other anticancer therapies in future clinical trials, in order to reverse acquired therapy resistance. Thus, epigenomics has the potential to generate relevant new knowledge of both a biological and clinical impact. Moreover, the potential, hurdles, and challenges of predictive biomarker discoveries will be discussed, with a special focus on the promise of liquid biopsies.
Collapse
Affiliation(s)
- Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia;
- Correspondence: ; Tel.: +421-2-3229-5198
| | - Julie Earl
- Molecular Epidemiology and Predictive Tumor Markers Group, Medical Oncology Research Laboratory, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain; (J.E.); (A.C.)
| | - María Laura García Bermejo
- Biomarkers and Therapeutic Targets Group, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain;
| | - Viola Stevurkova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia;
| | - Alfredo Carrato
- Molecular Epidemiology and Predictive Tumor Markers Group, Medical Oncology Research Laboratory, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain; (J.E.); (A.C.)
| | - Bozena Smolkova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia;
| |
Collapse
|
10
|
Gao S, Qin J, Sergeeva O, Sergeev M, Qiao P, Roelle S, Avril N, Lee Z, Li Y, Lu Z. Synthesis and assessment of ZD2-( 68Ga-NOTA) specific to extradomain B fibronectin in tumor microenvironment for PET imaging of pancreatic cancer. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2019; 9:216-229. [PMID: 31772820 PMCID: PMC6872477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/22/2019] [Indexed: 06/10/2023]
Abstract
Patients diagnosed with pancreatic cancer at a late stage have a dismal survival rate. Accurate early detection of pancreatic cancer with a size of 10 mm or less could dramatically improve patient survival after timely treatments. We have developed a new PET probe ZD2-(68Ga-NOTA) specific to extradomain-B fibronectin (EDB-FN), an oncoprotein in tumor microenvironment, for sensitive molecular imaging and early diagnosis of pancreatic cancer. A targeted ligand ZD2-NOTA is synthesized by conjugation of a macrocyclic ligand NOTA via a 6-aminohexanoic acid spacer to a linear ZD2 peptide (Thr-Val-Arg-Thr-Ser-Ala-Asp). ZD2-(68Ga-NOTA) is synthesized by relabeling of ZD2-NOTA with 68GaCl3 in a high purity under GMP conditions. The expression of EDB-FN is demonstrated in BxPC3 and Capan-1 human pancreatic cancer cells and tumor xenografts in mice. ZD2-(68Ga-NOTA) results in significantly higher uptake in the both BxPC3 and Capan-1 tumor xenografts than normal organs and tissues, including the brain, heart, liver and muscle, at 1 hr postinjection in mice. The tumor to muscle uptake ratio is at least 5 folds for the tracer in both tumors. ZD2-(68Ga-NOTA) is able to clearly delineate the PaCa tumors with a size of 10 mm or less with minimal background noise in normal tissues, including the liver. Substantial tumor uptake is still visible at 2 hr post-injection. The results suggest that the ZD2 peptide targeted PET probe has a potential for sensitive molecular imaging of EDB-FN and early detection of pancreatic cancer to improve healthcare of the patients diagnosed with the disease.
Collapse
Affiliation(s)
- Songqi Gao
- Department of Biomedical Engineering, Case Western Reserve UniversityCleveland, OH 44106, USA
- Molecular TheranosticsCleveland, OH 44114, USA
| | - Jingcan Qin
- Department of Biomedical Engineering, Case Western Reserve UniversityCleveland, OH 44106, USA
| | - Olga Sergeeva
- Department of Radiology, Case Western Reserve UniversityCleveland, OH 44106, USA
| | - Maxim Sergeev
- University Hospitals Cleveland Medical CenterCleveland, OH 44106, USA
| | - Peter Qiao
- Department of Biomedical Engineering, Case Western Reserve UniversityCleveland, OH 44106, USA
| | - Sarah Roelle
- Department of Biomedical Engineering, Case Western Reserve UniversityCleveland, OH 44106, USA
| | - Norbert Avril
- Department of Radiology, Case Western Reserve UniversityCleveland, OH 44106, USA
- University Hospitals Cleveland Medical CenterCleveland, OH 44106, USA
| | - Zhenghong Lee
- Department of Radiology, Case Western Reserve UniversityCleveland, OH 44106, USA
| | - Yajuan Li
- Molecular TheranosticsCleveland, OH 44114, USA
| | - Zhengrong Lu
- Department of Biomedical Engineering, Case Western Reserve UniversityCleveland, OH 44106, USA
- Department of Radiology, Case Western Reserve UniversityCleveland, OH 44106, USA
| |
Collapse
|
11
|
Fukamachi K, Hagiwara Y, Futakuchi M, Alexander DB, Tsuda H, Suzui M. Evaluation of a biomarker for the diagnosis of pancreas cancer using an animal model. J Toxicol Pathol 2019; 32:135-141. [PMID: 31404387 PMCID: PMC6682554 DOI: 10.1293/tox.2018-0062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/27/2019] [Indexed: 12/03/2022] Open
Abstract
Many approaches have been taken to identify new biomarkers of pancreatic ductal
carcinoma (PDC). Since animal models can be sampled under controlled conditions, better
standardization is possible compared with heterogeneous human studies. Transgenic rats
with conditional activation of oncogenic RAS in pancreatic tissue develop PDC that closely
resembles the biological and histopathological features of human PDC. Using this model, we
evaluated the usefulness of leucine-rich α2-glycoprotein-1 (LRG-1) as a serum marker. In
this study, we found that LRG-1 was overexpressed in rat PDC compared with normal pancreas
tissue of the control rats. Serum levels of LRG-1 were also significantly higher in rats
bearing PDC than in controls. Importantly, chronic pancreatitis in male Wistar Bonn/Kobori
rats, which is a widely accepted as a model of chronic pancreatitis, did not cause serum
levels of LRG-1 to become elevated. These results strongly support serum LRG-1 as a
candidate biomarker for noninvasive diagnosis of PDC. Our models of pancreas cancer
provide a useful strategy for evaluation of candidate markers applicable to human
cancer.
Collapse
Affiliation(s)
- Katsumi Fukamachi
- Department of Molecular Toxicology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Yoshiaki Hagiwara
- Immuno-Biological Laboratories, 1091-1 Naka, Fujioka-shi, Gunma 375-0005, Japan
| | - Mitsuru Futakuchi
- Department of Molecular Toxicology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - David B Alexander
- Nanotoxicology Project, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Hiroyuki Tsuda
- Nanotoxicology Project, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Masumi Suzui
- Department of Molecular Toxicology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| |
Collapse
|
12
|
Luchini C, Veronese N, Nottegar A, Cappelletti V, Daidone MG, Smith L, Parris C, Brosens LAA, Caruso MG, Cheng L, Wolfgang CL, Wood LD, Milella M, Salvia R, Scarpa A. Liquid Biopsy as Surrogate for Tissue for Molecular Profiling in Pancreatic Cancer: A Meta-Analysis Towards Precision Medicine. Cancers (Basel) 2019; 11:cancers11081152. [PMID: 31405192 PMCID: PMC6721631 DOI: 10.3390/cancers11081152] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 12/24/2022] Open
Abstract
Liquid biopsy (LB) is a non-invasive approach representing a promising tool for new precision medicine strategies for cancer treatment. However, a comprehensive analysis of its reliability for pancreatic cancer (PC) is lacking. To this aim, we performed the first meta-analysis on this topic. We calculated the pooled sensitivity, specificity, positive (LR+) and negative (LR−) likelihood ratio, and diagnostic odds ratio (DOR). A summary receiver operating characteristic curve (SROC) and area under curve (AUC) were used to evaluate the overall accuracy. We finally assessed the concordance rate of all mutations detected by multi-genes panels. Fourteen eligible studies involving 369 patients were included. The overall pooled sensitivity and specificity were 0.70 and 0.86, respectively. The LR+ was 3.85, the LR- was 0.34 and DOR was 15.84. The SROC curve with an AUC of 0.88 indicated a relatively high accuracy of LB for molecular characterization of PC. The concordance rate of all mutations detected by multi-genes panels was 31.9%. LB can serve as surrogate for tissue in the molecular profiling of PC, because of its relatively high sensitivity, specificity and accuracy. It represents a unique opportunity to be further explored towards its introduction in clinical practice and for developing new precision medicine approaches against PC.
Collapse
Affiliation(s)
- Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy
| | - Nicola Veronese
- National Institute of Gastroenterology-Research Hospital, IRCCS "S. de Bellis", Castellana Grotte, 70013 Bari, Italy
| | - Alessia Nottegar
- Department of Surgery, Section of Pathology, San Bortolo Hospital, 36100 Vicenza, Italy
| | - Vera Cappelletti
- Applied Research and Technological Development Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milano, Italy
| | - Maria G Daidone
- Applied Research and Technological Development Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milano, Italy
| | - Lee Smith
- Faculty of Science and Engineering, Anglia Ruskin University, Cambridge CB1 1PT, UK
| | - Christopher Parris
- Faculty of Science and Engineering, Anglia Ruskin University, Cambridge CB1 1PT, UK
| | - Lodewijk A A Brosens
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy
- Department of Pathology, University Medical Center Utrecht, Utrecht University, 3584CX Utrecht, The Netherlands
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6526GA Nijmegen, The Netherlands
| | - Maria G Caruso
- National Institute of Gastroenterology-Research Hospital, IRCCS "S. de Bellis", Castellana Grotte, 70013 Bari, Italy
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Christopher L Wolfgang
- Department of Surgery, Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21211, USA
| | - Laura D Wood
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21211, USA
- Department of Oncology, Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21211, USA
| | - Michele Milella
- Department of Medicine, Section of Medical Oncology, University and Hospital Trust of Verona, 37134 Verona, Italy
| | - Roberto Salvia
- Department of General and Visceral Surgery, The Pancreas Institute, University and Hospital Trust of Verona, 37134 Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy.
- ARC-Net Research Center, University of Verona, 37134 Verona, Italy.
| |
Collapse
|
13
|
Buscail E, Maulat C, Muscari F, Chiche L, Cordelier P, Dabernat S, Alix-Panabières C, Buscail L. Liquid Biopsy Approach for Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2019; 11:cancers11060852. [PMID: 31248203 PMCID: PMC6627808 DOI: 10.3390/cancers11060852] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/01/2019] [Accepted: 06/14/2019] [Indexed: 01/10/2023] Open
Abstract
Pancreatic cancer is a public health problem because of its increasing incidence, the absence of early diagnostic tools, and its aggressiveness. Despite recent progress in chemotherapy, the 5-year survival rate remains below 5%. Liquid biopsies are of particular interest from a clinical point of view because they are non-invasive biomarkers released by primary tumours and metastases, remotely reflecting disease burden. Pilot studies have been conducted in pancreatic cancer patients evaluating the detection of circulating tumour cells, cell-free circulating tumour DNA, exosomes, and tumour-educated platelets. There is heterogeneity between the methods used to isolate circulating tumour elements as well as the targets used for their identification. Performances for the diagnosis of pancreatic cancer vary depending of the technique but also the stage of the disease: 30–50% of resectable tumours are positive and 50–100% are positive in locally advanced and/or metastatic cases. A significant prognostic value is demonstrated in 50–70% of clinical studies, irrespective of the type of liquid biopsy. Large prospective studies of homogeneous cohorts of patients are lacking. One way to improve diagnostic and prognostic performances would be to use a combined technological approach for the detection of circulating tumour cells, exosomes, and DNA.
Collapse
Affiliation(s)
- Etienne Buscail
- INSERM U1035, Bordeaux University, 33000 Bordeaux, France.
- Department of Digestive Surgery, Bordeaux University Hospital, 33600 Pessac, France.
| | - Charlotte Maulat
- Université Fédérale Toulouse Midi-Pyrénées, Université Toulouse III Paul Sabatier, INSERM, CRCT, 31330 Toulouse, France.
- Department of Digestive Surgery, Toulouse University Hospital, 31059 Toulouse, France.
| | - Fabrice Muscari
- Université Fédérale Toulouse Midi-Pyrénées, Université Toulouse III Paul Sabatier, INSERM, CRCT, 31330 Toulouse, France.
- Department of Digestive Surgery, Toulouse University Hospital, 31059 Toulouse, France.
| | - Laurence Chiche
- INSERM U1035, Bordeaux University, 33000 Bordeaux, France.
- Department of Digestive Surgery, Bordeaux University Hospital, 33600 Pessac, France.
| | - Pierre Cordelier
- Université Fédérale Toulouse Midi-Pyrénées, Université Toulouse III Paul Sabatier, INSERM, CRCT, 31330 Toulouse, France.
| | | | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), Montpellier Hospital and University of Montpellier, 34295 Montpellier, France.
| | - Louis Buscail
- Université Fédérale Toulouse Midi-Pyrénées, Université Toulouse III Paul Sabatier, INSERM, CRCT, 31330 Toulouse, France.
- Department of Gastroenterology and Pancreatology, Toulouse University Hospital, 31059 Toulouse, France.
| |
Collapse
|
14
|
Abstract
Since its discovery in human blood plasma about 70 years ago, circulating cell-free DNA (cfDNA) has become an attractive subject of research as noninvasive disease biomarker. The interest in clinical applications has gained an exponential increase, making it a popular and potential target in a wide range of research areas.cfDNA can be found in different body fluids, both in healthy and not healthy subjects. The recent and rapid development of new molecular techniques is promoting the study and the identification of cfDNA, holding the key to minimally invasive diagnostics, improving disease monitoring, clinical decision, and patients' outcome.cfDNA has already given a huge impact on prenatal medicine, and it could become, in the next future, the standard of care also in other fields, from oncology to transplant medicine and cardiovascular diseases.
Collapse
Affiliation(s)
- Rossella Ranucci
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy.
| |
Collapse
|
15
|
|
16
|
Prognostic stratification of resected pancreatic ductal adenocarcinoma: Past, present, and future. Dig Liver Dis 2018; 50:979-990. [PMID: 30205952 DOI: 10.1016/j.dld.2018.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/02/2018] [Accepted: 08/02/2018] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the digestive cancer with the poorest prognosis, with a 5-year overall survival rate of 7%. Complete surgical resection followed by adjuvant chemotherapy is the only treatment with curative intent. However, many patients with an apparently localized disease who may undergo primary tumor resection already have micro-metastatic disease and will promptly develop metastases. Considering the significant rate of morbidity and mortality upon pancreatic surgery, the pre-operative identification of patients with an aggressive disease is therefore a major clinical issue. Although tumor size, differentiation, margins, and lymph node invasion are the main "classical" prognostic factors, they are not sufficient to fully predict early disease recurrence. In the last decade, multi-omics high-throughput analyses have provided a new insight into PDAC biology and have led to the description of multiple molecular subtypes, with a significant prognostic value for most of them, but that have not yet been transposed to routine clinical practice, mainly due to poor availability of tumor tissue material prior to surgical resection. In this review, we provide an overview of the current status of clinico-pathological and molecular biomarkers (tumor and blood) to predict early recurrence, and their implications for clinical practice and future research development.
Collapse
|
17
|
Kunovsky L, Tesarikova P, Kala Z, Kroupa R, Kysela P, Dolina J, Trna J. The Use of Biomarkers in Early Diagnostics of Pancreatic Cancer. Can J Gastroenterol Hepatol 2018; 2018:5389820. [PMID: 30186820 PMCID: PMC6112218 DOI: 10.1155/2018/5389820] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/27/2018] [Accepted: 08/06/2018] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid malignancies with increasing incidence. The poor prognosis is due to the aggressive nature of the tumor, late detection, and the resistance to chemotherapy and radiotherapy. A radical surgery procedure is the only treatment that has been shown to improve the 5-year survival rate to 20-25%. However, the majority of patients (80-85%) are diagnosed with locally advanced or metastatic disease and just 15-20% patients are diagnosed in an early stage allowing them to undergo the potentially curative surgical resection. The early detection of PDAC without the use of invasive methods is challenging and discovery of a cost-effective biomarker with high specificity and sensitivity could significantly improve the treatment and survival in these patients. In this review, we summarize current and newly examined biomarkers in early PDAC detection.
Collapse
Affiliation(s)
- Lumir Kunovsky
- Department of Gastroenterology, University Hospital Brno Bohunice, Faculty of Medicine, Masaryk University, Czech Republic
- Department of Surgery, University Hospital Brno Bohunice, Faculty of Medicine, Masaryk University, Czech Republic
| | - Pavla Tesarikova
- Department of Internal Medicine, Hospital Boskovice, Czech Republic
| | - Zdenek Kala
- Department of Surgery, University Hospital Brno Bohunice, Faculty of Medicine, Masaryk University, Czech Republic
| | - Radek Kroupa
- Department of Gastroenterology, University Hospital Brno Bohunice, Faculty of Medicine, Masaryk University, Czech Republic
| | - Petr Kysela
- Department of Surgery, University Hospital Brno Bohunice, Faculty of Medicine, Masaryk University, Czech Republic
| | - Jiri Dolina
- Department of Gastroenterology, University Hospital Brno Bohunice, Faculty of Medicine, Masaryk University, Czech Republic
| | - Jan Trna
- Department of Gastroenterology, University Hospital Brno Bohunice, Faculty of Medicine, Masaryk University, Czech Republic
- Department of Internal Medicine, Hospital Boskovice, Czech Republic
| |
Collapse
|
18
|
Domínguez-Vigil IG, Moreno-Martínez AK, Wang JY, Roehrl MH, Barrera-Saldaña HA. The dawn of the liquid biopsy in the fight against cancer. Oncotarget 2018; 9:2912-2922. [PMID: 29416824 PMCID: PMC5788692 DOI: 10.18632/oncotarget.23131] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 11/10/2017] [Indexed: 01/06/2023] Open
Abstract
Cancer is a molecular disease associated with alterations in the genome, which, thanks to the highly improved sensitivity of mutation detection techniques, can be identified in cell-free DNA (cfDNA) circulating in blood, a method also called liquid biopsy. This is a non-invasive alternative to surgical biopsy and has the potential of revealing the molecular signature of tumors to aid in the individualization of treatments. In this review, we focus on cfDNA analysis, its advantages, and clinical applications employing genomic tools (NGS and dPCR) particularly in the field of oncology, and highlight its valuable contributions to early detection, prognosis, and prediction of treatment response.
Collapse
Affiliation(s)
- Irma G. Domínguez-Vigil
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina de la Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | - Ana K. Moreno-Martínez
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina de la Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
- Genetics Laboratory, Vitagénesis, Monterrey, Nuevo León, México
| | | | - Michael H.A. Roehrl
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hugo A. Barrera-Saldaña
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina de la Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
- TecSalud, Tecnológico de Monterrey, San Pedro Garza García, Nuevo León, México
| |
Collapse
|
19
|
Adamo P, Cowley CM, Neal CP, Mistry V, Page K, Dennison AR, Isherwood J, Hastings R, Luo J, Moore DA, Howard PJ, Miguel ML, Pritchard C, Manson M, Shaw JA. Profiling tumour heterogeneity through circulating tumour DNA in patients with pancreatic cancer. Oncotarget 2017; 8:87221-87233. [PMID: 29152076 PMCID: PMC5675628 DOI: 10.18632/oncotarget.20250] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/14/2017] [Indexed: 01/17/2023] Open
Abstract
The majority of pancreatic ductal adenocarcinomas (PDAC) are diagnosed late so that surgery is rarely curative. Earlier detection could significantly increase the likelihood of successful treatment and improve survival. The aim of the study was to provide proof of principle that point mutations in key cancer genes can be identified by sequencing circulating free DNA (cfDNA) and that this could be used to detect early PDACs and potentially, premalignant lesions, to help target early effective treatment. Targeted next generation sequencing (tNGS) analysis of mutation hotspots in 50 cancer genes was conducted in 26 patients with PDAC, 14 patients with chronic pancreatitis (CP) and 12 healthy controls with KRAS status validated by digital droplet PCR. A higher median level of total cfDNA was observed in patients with PDAC (585 ng/ml) compared to either patients with CP (300 ng/ml) or healthy controls (175 ng/ml). PDAC tissue showed wide mutational heterogeneity, whereas KRAS was the most commonly mutated gene in cfDNA of patients with PDAC and was significantly associated with a poor disease specific survival (p=0.018). This study demonstrates that tNGS of cfDNA is feasible to characterise the circulating genomic profile in PDAC and that driver mutations in KRAS have prognostic value but cannot currently be used to detect early emergence of disease. Importantly, monitoring total cfDNA levels may have utility in individuals "at risk" and warrants further investigation.
Collapse
Affiliation(s)
- Patricia Adamo
- Department of Cancer Studies, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, UK
| | - Caroline M Cowley
- Department of Cancer Studies, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, UK
| | - Christopher P Neal
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - Vilas Mistry
- Department of Cancer Studies, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, UK
| | - Karen Page
- Department of Cancer Studies, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, UK
| | - Ashley R Dennison
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - John Isherwood
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - Robert Hastings
- Cancer Research UK Leicester Centre, University of Leicester, Leicester, UK
| | - JinLi Luo
- Cancer Research UK Leicester Centre, University of Leicester, Leicester, UK
| | - David A Moore
- Department of Cancer Studies, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, UK
| | - Pringle J Howard
- Department of Cancer Studies, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, UK
| | | | - Catrin Pritchard
- Department of Cancer Studies, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, UK
| | - Margaret Manson
- Department of Cancer Studies, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, UK
| | - Jacqui A Shaw
- Department of Cancer Studies, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, UK
| |
Collapse
|
20
|
Segelov E, Lordick F, Goldstein D, Chantrill LA, Croagh D, Lawrence B, Arnold D, Chau I, Obermannova R, Price TJ. Current challenges in optimizing systemic therapy for patients with pancreatic cancer: expert perspectives from the Australasian Gastrointestinal Trials Group (AGITG) with invited international faculty. Expert Rev Anticancer Ther 2017; 17:951-964. [PMID: 28817982 DOI: 10.1080/14737140.2017.1369882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Despite recent progress, the outlook for most patients with pancreatic cancer remains poor. There is variation in how patients are managed globally due to differing interpretations of the evidence, partly because studies in this disease are challenging to undertake. This article collates the evidence upon which current best practice is based and offers an expert opinion from an international faculty on how latest developments should influence current treatment paradigms. Areas covered: Optimal chemotherapy for first and subsequent lines of therapy; optimal management of locally advanced, non-metastatic cancer including the role of neoadjuvant chemo(radio)therapy, current evidence for adjuvant chemotherapy, major advances in pancreatic cancer genomics and challenges in supportive care particularly relevant to patients with pancreatic cancer. For each section, literature was reviewed by comprehensive search techniques, including clinical trial websites and abstracts from international cancer meetings. Expert commentary: For each section, a commentary is provided. Overall the challenges identified were: difficulties in diagnosing pancreatic cancer early, challenges for performing randomised clinical trials in all stages of the disease, some progress in systemic therapy with new agents and in identifying molecular subtypes that may be clinically relevant and move towards personalized therapy, but still, pancreatic cancer remains a very poor prognosis cancer with significant palliative care needs.
Collapse
Affiliation(s)
- Eva Segelov
- a Department of Oncology , Monash Medical Centre and Monash University , Melbourne , Australia
| | - Florian Lordick
- b Department of Oncology, University Cancer Center Leipzig , University Medicine Leipzig , Leipzig , Germany
| | - David Goldstein
- c Department of Oncology, Nelune Cancer Centre , Prince of Wales Hospital and University of New South Wales , Sydney , Australia
| | - Lorraine A Chantrill
- d Department of Oncology , The Kinghorn Cancer Centre and University of Western Sydney , Sydney , Australia
| | - Daniel Croagh
- a Department of Oncology , Monash Medical Centre and Monash University , Melbourne , Australia
| | - Ben Lawrence
- e Department of Oncology , University of Auckland , Auckland , New Zealand
| | - Dirk Arnold
- f Department of Oncology , Instituto CUF de Oncologia , Lisbon , Portugal
| | - Ian Chau
- g Department of Oncology , Royal Marsden Hospital , London & Surrey , UK
| | - Radka Obermannova
- h Department of Comprehensive Cancer Care , Masaryk Memorial Cancer Institute , Brno , Czech Republic
| | - Timothy Jay Price
- i Queen Elizabeth Hospital and Lyell McEwin Hospital , Adelaide , Australia
| |
Collapse
|
21
|
Moravec R, Divi R, Verma M. Detecting circulating tumor material and digital pathology imaging during pancreatic cancer progression. World J Gastrointest Oncol 2017; 9:235-250. [PMID: 28656074 PMCID: PMC5472554 DOI: 10.4251/wjgo.v9.i6.235] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/04/2017] [Accepted: 03/24/2017] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer (PC) is a leading cause of cancer-related death worldwide. Clinical symptoms typically present late when treatment options are limited and survival expectancy is very short. Metastatic mutations are heterogeneous and can accumulate up to twenty years before PC diagnosis. Given such genetic diversity, detecting and managing the complex states of disease progression may be limited to imaging modalities and markers present in circulation. Recent developments in digital pathology imaging show potential for early PC detection, making a differential diagnosis, and predicting treatment sensitivity leading to long-term survival in advanced stage patients. Despite large research efforts, the only serum marker currently approved for clinical use is CA 19-9. Utility of CA 19-9 has been shown to improve when it is used in combination with PC-specific markers. Efforts are being made to develop early-screening assays that can detect tumor-derived material, present in circulation, before metastasis takes a significant course. Detection of markers that identify circulating tumor cells and tumor-derived extracellular vesicles (EVs) in biofluid samples offers a promising non-invasive method for this purpose. Circulating tumor cells exhibit varying expression of epithelial and mesenchymal markers depending on the state of tumor differentiation. This offers a possibility for monitoring disease progression using minimally invasive procedures. EVs also offer the benefit of detecting molecular cargo of tumor origin and add the potential to detect circulating vesicle markers from tumors that lack invasive properties. This review integrates recent genetic insights of PC progression with developments in digital pathology and early detection of tumor-derived circulating material.
Collapse
|