1
|
Hu K, Hasegawa K, Zhou G. Bioinformatics-based screening of key genes associated with gemcitabine resistance in advanced pancreatic ductal adenocarcinoma. Transl Cancer Res 2024; 13:6947-6955. [PMID: 39816538 PMCID: PMC11730203 DOI: 10.21037/tcr-2024-2374] [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: 11/27/2024] [Accepted: 12/12/2024] [Indexed: 01/18/2025]
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) ranks among the deadliest cancers globally. Despite gemcitabine being a primary chemotherapeutic agent, many patients with PDAC develop resistance, significantly limiting treatment efficacy. This study aims to screen and validate key genes associated with gemcitabine resistance in advanced PDAC using bioinformatics analysis and clinical sample validation, thereby providing potential noninvasive biomarkers and therapeutic targets for overcoming chemoresistance. Methods This study used bioinformatics approaches to analyze gene expression data from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases, identifying differentially expressed genes (DEGs) associated with gemcitabine resistance in advanced PDAC. A total of 122 patients with advanced PDAC were selected for the study and divided into gemcitabine-sensitive and gemcitabine-resistant groups post-treatment. The expression levels of key genes in patients' serum were measured using enzyme-linked immunosorbent assay, and both univariate and multivariate analyses were performed to assess their potential as noninvasive biomarkers for predicting resistance. Results Ten upregulated DEGs related to gemcitabine resistance were identified. Among these genes, cathepsin E (CTSE) was significantly negatively correlated with overall survival, disease-specific survival, and progression-free interval in patients with PDAC and was thus identified as a significant key gene. Further clinical sample validation confirmed that CTSE expression level was significantly higher in the resistant group of patients with advanced PDAC compared to the sensitive group, establishing CTSE as an independent predictor of gemcitabine resistance. Conclusions CTSE is a key gene associated with gemcitabine resistance in advanced PDAC and shows promise as a target for enhancing responsiveness to gemcitabine treatment.
Collapse
Affiliation(s)
- Kaifeng Hu
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Kiyoshi Hasegawa
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Guozhi Zhou
- Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
2
|
Wu Y, Zhou W, Yang Z, Li J, Jin Y. miR-185-5p Represses Cells Growth and Metastasis of Osteosarcoma via Targeting Cathepsin E. Int J Toxicol 2022; 41:115-125. [PMID: 35213250 DOI: 10.1177/10915818211069270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Osteosarcoma (OS) is a malignant bone tumor characterized by poor prognosis due to its regional invasion and early metastasis. In this study, we aimed to find the role and the underlying mechanism of Cathepsin E (CTSE) in OS growth and metastasis. We found CTSE is upregulated in metastatic OS, rather than in the primary lesion, as confirmed by RT-qPCR and western blot analysis of clinical OS samples. Furthermore, both in vitro and in vivo experiments illustrated that CTSE promoted both growth and metastasis of OS cells, partially mediated through the modulation of Epithelial-Mesenchymal Transition (EMT). Bioinformatics analysis predicted that miR-185-5p downregulates CTSE via directly binding to the 3'UTR of CTSE, which was verified by luciferase reporter assay and rescue assays. This study reported for the first time that CTSE is a potential biomarker in OS tumorigenesis and metastasis, providing a promising therapeutic target for OS treatment.
Collapse
Affiliation(s)
- Yue Wu
- Department of Orthopaedics, Beijing United Family Healthcare, Beijing, China
| | - Weili Zhou
- Joint Surgery Department of Orthopaedics, The Third Hospital of Changsha, Changsha, China
| | - Zhijun Yang
- Trauma Department of Orthopaedics, The First Affiliated Hospital of South China University, Hengyang, China
| | - Jinping Li
- Joint Surgery Department of Orthopaedics, Changsha Central Hospital, Changsha, China
| | - Yi Jin
- Joint Surgery Department of Orthopaedics, Changsha Central Hospital, Changsha, China
| |
Collapse
|
3
|
van Dam MA, Vuijk FA, Stibbe JA, Houvast RD, Luelmo SAC, Crobach S, Shahbazi Feshtali S, de Geus-Oei LF, Bonsing BA, Sier CFM, Kuppen PJK, Swijnenburg RJ, Windhorst AD, Burggraaf J, Vahrmeijer AL, Mieog JSD. Overview and Future Perspectives on Tumor-Targeted Positron Emission Tomography and Fluorescence Imaging of Pancreatic Cancer in the Era of Neoadjuvant Therapy. Cancers (Basel) 2021; 13:6088. [PMID: 34885196 PMCID: PMC8656821 DOI: 10.3390/cancers13236088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/25/2021] [Accepted: 11/28/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Despite recent advances in the multimodal treatment of pancreatic ductal adenocarcinoma (PDAC), overall survival remains poor with a 5-year cumulative survival of approximately 10%. Neoadjuvant (chemo- and/or radio-) therapy is increasingly incorporated in treatment strategies for patients with (borderline) resectable and locally advanced disease. Neoadjuvant therapy aims to improve radical resection rates by reducing tumor mass and (partial) encasement of important vascular structures, as well as eradicating occult micrometastases. Results from recent multicenter clinical trials evaluating this approach demonstrate prolonged survival and increased complete surgical resection rates (R0). Currently, tumor response to neoadjuvant therapy is monitored using computed tomography (CT) following the RECIST 1.1 criteria. Accurate assessment of neoadjuvant treatment response and tumor resectability is considered a major challenge, as current conventional imaging modalities provide limited accuracy and specificity for discrimination between necrosis, fibrosis, and remaining vital tumor tissue. As a consequence, resections with tumor-positive margins and subsequent early locoregional tumor recurrences are observed in a substantial number of patients following surgical resection with curative intent. Of these patients, up to 80% are diagnosed with recurrent disease after a median disease-free interval of merely 8 months. These numbers underline the urgent need to improve imaging modalities for more accurate assessment of therapy response and subsequent re-staging of disease, thereby aiming to optimize individual patient's treatment strategy. In cases of curative intent resection, additional intra-operative real-time guidance could aid surgeons during complex procedures and potentially reduce the rate of incomplete resections and early (locoregional) tumor recurrences. In recent years intraoperative imaging in cancer has made a shift towards tumor-specific molecular targeting. Several important molecular targets have been identified that show overexpression in PDAC, for example: CA19.9, CEA, EGFR, VEGFR/VEGF-A, uPA/uPAR, and various integrins. Tumor-targeted PET/CT combined with intraoperative fluorescence imaging, could provide valuable information for tumor detection and staging, therapy response evaluation with re-staging of disease and intraoperative guidance during surgical resection of PDAC. METHODS A literature search in the PubMed database and (inter)national trial registers was conducted, focusing on studies published over the last 15 years. Data and information of eligible articles regarding PET/CT as well as fluorescence imaging in PDAC were reviewed. Areas covered: This review covers the current strategies, obstacles, challenges, and developments in targeted tumor imaging, focusing on the feasibility and value of PET/CT and fluorescence imaging for integration in the work-up and treatment of PDAC. An overview is given of identified targets and their characteristics, as well as the available literature of conducted and ongoing clinical and preclinical trials evaluating PDAC-targeted nuclear and fluorescent tracers.
Collapse
Affiliation(s)
- Martijn A. van Dam
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Floris A. Vuijk
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Judith A. Stibbe
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Ruben D. Houvast
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Saskia A. C. Luelmo
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Stijn Crobach
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | | | - Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, University Medical Center Leiden, 2333 ZA Leiden, The Netherlands;
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, The Netherlands
| | - Bert A. Bonsing
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Cornelis F. M. Sier
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
- Percuros B.V., 2333 CL Leiden, The Netherlands
| | - Peter J. K. Kuppen
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | | | - Albert D. Windhorst
- Department of Radiology, Section of Nuclear Medicine, Amsterdam UMC, Location VUmc, 1081 HV Amsterdam, The Netherlands;
| | - Jacobus Burggraaf
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
- Centre for Human Drug Research, 2333 CL Leiden, The Netherlands
| | - Alexander L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - J. Sven D. Mieog
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| |
Collapse
|
4
|
Takahashi R, Ishizawa T, Sato M, Inagaki Y, Takanka M, Kuriki Y, Kamiya M, Ushiku T, Urano Y, Hasegawa K. Fluorescence Imaging Using Enzyme-Activatable Probes for Real-Time Identification of Pancreatic Cancer. Front Oncol 2021; 11:714527. [PMID: 34490111 PMCID: PMC8417470 DOI: 10.3389/fonc.2021.714527] [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: 05/25/2021] [Accepted: 08/04/2021] [Indexed: 01/11/2023] Open
Abstract
Introduction Radical resection is the only curative treatment for pancreatic cancer, which is a life-threatening disease. However, it is often not easy to accurately identify the extent of the tumor before and during surgery. Here we describe the development of a novel method to detect pancreatic tumors using a tumor-specific enzyme-activatable fluorescence probe. Methods Tumor and non-tumor lysate or small specimen collected from the resected specimen were selected to serve as the most appropriate fluorescence probe to distinguish cancer tissues from noncancerous tissues. The selected probe was sprayed onto the cut surface of the resected specimen of cancer tissue to acquire a fluorescence image. Next, we evaluated the ability of the probe to detect the tumor and calculated the tumor-to-background ratio (TBR) by comparing the fluorescence image with the pathological extent of the tumor. Finally, we searched for a tumor-specific enzyme that optimally activates the selected probe. Results Using a library comprising 309 unique fluorescence probes, we selected GP-HMRG as the most appropriate activatable fluorescence probe. We obtained eight fluorescence images of resected specimens, among which four approximated the pathological findings of the tumor, which achieved the highest TBR. Finally, dipeptidyl-peptidase IV (DPP-IV) or a DPP-IV-like enzyme was identified as the target enzyme. Conclusion This novel method may enable rapid and real-time visualization of pancreatic cancer through the enzymatic activities of cancer tissues.
Collapse
Affiliation(s)
- Ryugen Takahashi
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Takeaki Ishizawa
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Masumitsu Sato
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yoshinori Inagaki
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Mariko Takanka
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yugo Kuriki
- Laboratory of Chemistry and Biology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Mako Kamiya
- Laboratory of Chemical Biology and Molecular Imaging, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuteru Urano
- Laboratory of Chemistry and Biology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,Laboratory of Chemical Biology and Molecular Imaging, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Hasegawa
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Jugniot N, Bam R, Meuillet EJ, Unger EC, Paulmurugan R. Current status of targeted microbubbles in diagnostic molecular imaging of pancreatic cancer. Bioeng Transl Med 2021; 6:e10183. [PMID: 33532585 PMCID: PMC7823123 DOI: 10.1002/btm2.10183] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/19/2020] [Accepted: 08/19/2020] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is often associated with a poor prognosis due to silent onset, resistance to therapies, and rapid spreading. Most patients are ineligible for curable surgery as they present with advanced disease at the time of diagnosis. Present diagnostic methods relying on anatomical changes have various limitations including difficulty to discriminate between benign and malignant conditions, invasiveness, the ambiguity of imaging results, or the inability to detect molecular biomarkers of PDAC initiation and progression. Therefore, new imaging technologies with high sensitivity and specificity are critically needed for accurately detecting PDAC and noninvasively characterizing molecular features driving its pathogenesis. Contrast enhanced targeted ultrasound (CETUS) is an upcoming molecular imaging modality that specifically addresses these issues. Unlike anatomical imaging modalities such as CT and MRI, molecular imaging using CETUS is promising for early and accurate detection of PDAC. The use of molecularly targeted microbubbles that bind to neovascular targets can enhance the ultrasound signal specifically from malignant PDAC tissues. This review discusses the current state of diagnostic imaging modalities for pancreatic cancer and places a special focus on ultrasound targeted-microbubble technology together with its clinical translatability for PDAC detection.
Collapse
Affiliation(s)
- Natacha Jugniot
- Department of RadiologyMolecular Imaging Program at Stanford, Stanford UniversityPalo AltoCaliforniaUSA
| | - Rakesh Bam
- Department of RadiologyMolecular Imaging Program at Stanford, Stanford UniversityPalo AltoCaliforniaUSA
| | | | | | - Ramasamy Paulmurugan
- Department of RadiologyMolecular Imaging Program at Stanford, Stanford UniversityPalo AltoCaliforniaUSA
| |
Collapse
|
6
|
Castellani D, Germani U, Bassotti G, Manta R. EUS Standard Devices. ENDOTHERAPY IN BILIOPANCREATIC DISEASES: ERCP MEETS EUS 2020:47-63. [DOI: 10.1007/978-3-030-42569-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
7
|
Pontious C, Kaul S, Hong M, Hart PA, Krishna SG, Lara L, Conwell DL, Cruz-Monserrate Z. Cathepsin E expression and activity: Role in the detection and treatment of pancreatic cancer. Pancreatology 2019; 19:951-956. [PMID: 31582345 PMCID: PMC6829043 DOI: 10.1016/j.pan.2019.09.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 09/19/2019] [Indexed: 12/11/2022]
Abstract
Cathepsin E (CTSE) is an intracellular, hydrolytic aspartic protease found to be expressed in cells of the immune and gastrointestinal systems, lymphoid tissues, erythrocytes, and cancer cells. The precise functions are not fully understood; however, various studies have investigated its numerous cell-type specific roles. CTSE expression has been shown to be a potential early biomarker for pancreatic ductal adenocarcinoma (PDAC). PDAC patients have low survival rates mostly due to the lack of early detection methods. CTSE-specific activity probes have been developed and tested to assist in tumor imaging and functional studies investigating the role of CTSE expression in PDAC tumors. Furthermore, a CTSE protease-specific, photodynamic therapy pro-drug was developed to explore its potential use to treat tumors that express CTSE. Since CTSE is expressed in pancreatic diseases that are risk factors for PDAC, such as pancreatic cysts and chronic pancreatitis, learning about its function in these disease types could assist in early PDAC detection and in understanding the biology of PDAC progression. Overall, CTSE expression and activity shows potential to detect PDAC and other pancreatic diseases. Further research is needed to fully understand its functions and potential translational applicability.
Collapse
Affiliation(s)
- Corbin Pontious
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH,The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Sabrina Kaul
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH,The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Marcus Hong
- The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH,Kenyon College, Gambier, OH
| | - Phil A. Hart
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Somashekar G. Krishna
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH,The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Luis Lara
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Darwin L. Conwell
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH,The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH,The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| |
Collapse
|
8
|
The clinical evaluation of needle-based confocal laser endomicroscopy in the assessment of pancreatic cystic lesion: A pilot study. PROCEEDINGS OF SINGAPORE HEALTHCARE 2018. [DOI: 10.1177/2010105817731798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background/aim: Pancreatic cystic lesions are increasingly diagnosed from cross-sectional imaging done for other indications. The challenge lies in the ability to correctly identify the high-risk individuals for resection surgery, which carries high morbidity and mortality. Unfortunately, present diagnostic techniques are suboptimal. Needle-based confocal laser endomicroscopy (nCLE) has been designed to bridge this diagnostic gap. We aim to assess the feasibility and safety of nCLE in the assessment of pancreatic cystic lesions. Methods: We prospectively recruited patients referred for assessment of pancreatic cystic lesions from August 2014 until July 2015. All pancreatic cystic lesions were examined with nCLE miniprobe via endoscopic ultrasound followed by fine-needle aspiration. Information regarding the cysts (morphology, location, fluid analysis etc.) was documented. Adverse event was recorded. Results: Fourteen patients were recruited, six were male with a mean age of 66.5 (range 48–80) years. Only 12 completed nCLE examination of the pancreatic cystic lesions. nCLE imaging was successful in 83.3% (10/12). Average nCLE imaging duration was 5 min 18 s. There was one (10%) adverse event. nCLE impressions were correct in eight cases (five malignant and three benign) compared with final diagnosis. Three patients underwent surgery, histology showed ductal adenocarcinoma, pancreatic neuroendocrine tumor and gastric-subtype of intraductal papillary mucinous neoplasm. The sensitivity and specificity of the nCLE impression when compared with final diagnosis were 83.3% and 75% respectively. The accuracy of nCLE was 80%. Conclusion: Our results have demonstrated that nCLE assessment of pancreatic cystic lesions is safe and feasible. It may complement the existing diagnostic modalities to improve diagnostic yield.
Collapse
|
9
|
Cazacu IM, Luzuriaga Chavez AA, Saftoiu A, Vilmann P, Bhutani MS. A quarter century of EUS-FNA: Progress, milestones, and future directions. Endosc Ultrasound 2018; 7:141-160. [PMID: 29941723 PMCID: PMC6032705 DOI: 10.4103/eus.eus_19_18] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 03/21/2018] [Indexed: 12/11/2022] Open
Abstract
Tissue acquisition using EUS has considerably evolved since the first EUS-FNA was reported 25 years ago. Its introduction was an important breakthrough in the endoscopic field. EUS-FNA has now become a part of the diagnostic and staging algorithm for the evaluation of benign and malignant diseases of the gastrointestinal tract and of the organs in its proximity, including lung diseases. This review aims to present the history of EUS-FNA development and to provide a perspective on the recent developments in procedural techniques and needle technologies that have significantly extended the role of EUS and its clinical applications. There is a bright future ahead for EUS-FNA in the years to come as extensive research is conducted in this field and various technologies are continuously implemented into clinical practice.
Collapse
Affiliation(s)
- Irina Mihaela Cazacu
- Department of Gastroenterology, Research Center of Gastroenterology and Hepatology, University of Medicine and Pharmacy, Craiova, Romania
- Department of Gastroenterology, Hepatology, and Nutrition, University of Texas – MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Adrian Saftoiu
- Department of Gastroenterology, Research Center of Gastroenterology and Hepatology, University of Medicine and Pharmacy, Craiova, Romania
| | - Peter Vilmann
- Gastrounit, Division of Surgery, Copenhagen University Hospital Herlev, Copenhagen, Denmark
| | - Manoop S. Bhutani
- Department of Gastroenterology, Hepatology, and Nutrition, University of Texas – MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
10
|
Abstract
Cathepsins are lysosomal peptidases belonging to the papain family, and based on their catalytic sites, these enzymes can be divided into serine, cysteine and aspartic proteases. The studies conducted to date have identified, 15 types of cathepsins that are widely distributed in intracellular and extracellular spaces. These proteases participate in various pathological activities, including the occurrence and development of human cancers. Several recent studies suggest that cathepsins, particularly cathepsins B, D, E and L, contribute to digestive tumorigenesis. Cathepsins were found to promote the development of most digestive cancers except liver cancer, in which they might have the opposite effects. Due to their important roles in digestive tumors, cathepsins might be therapeutic targets for the treatment of digestive cancers.
Collapse
|
11
|
Li H, Wang P, Gong W, Wang Q, Zhou J, Zhu WH, Cheng Y. Dendron-Grafted Polylysine-Based Dual-Modal Nanoprobe for Ultra-Early Diagnosis of Pancreatic Precancerosis via Targeting a Urokinase-Type Plasminogen Activator Receptor. Adv Healthc Mater 2018; 7. [PMID: 29195018 DOI: 10.1002/adhm.201700912] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/17/2017] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer death. Early detection of precancerous pancreatic intraepithelial neoplasia (PanIN) tissues is an urgent challenge to improve the PDAC prognosis. Here, a urokinase-type plasminogen activator receptor (uPAR)-targeted magnetic resonance (MR)/near-infrared fluorescence (NIRF) dual-modal nanoprobe dendron-grafted polylysine (DGL)-U11 for ultra-early detection of pancreatic precancerosis is reported. Because of its good biocompatibility and biodegradability, globular architecture, and well-defined reactive groups, the DGL is chosen as the platform to load with a pancreatic tumor-targeting peptide U11, a magnetic resonance contrast agent Gd3+ -diethylene triamine pentaacetic acid, and a near-infrared fluorescent cyanine dye Cy5.5. The nanoprobe DGL-U11 has several preferable characteristics, such as active peptide targeting to activator receptor, good biocompatibility, dual-modal imaging diagnosis, and well controlled diameter in a range of 15-25 nm. Upon incorporation of the active U11 peptide target to the overexpressed activator receptor uPAR, the targeted nanoprobe DGL-U11 can increase to the earlier PanIN-II stage through in vivo NIRF imaging. Labeled with both MR and NIRF bioimaging reporters, the uPAR-targeted dual-modal nanoprobe is very effective in the targeted imaging of precancerous PanINs and PDAC lesions with high sensitivity and spatial resolution, providing a promising platform to the ultra-early detection of PDAC.
Collapse
Affiliation(s)
- Hui Li
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yi Shan Road, Shanghai, 200233, P. R. China
| | - Ping Wang
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Wenyu Gong
- Department of CT, the First People's Hospital of Yancheng City, Jiangsu, 224005, China
| | - Qi Wang
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals, Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Jia Zhou
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yi Shan Road, Shanghai, 200233, P. R. China
| | - Wei-Hong Zhu
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals, Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Yingsheng Cheng
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yi Shan Road, Shanghai, 200233, P. R. China
| |
Collapse
|
12
|
Combination of active targeting, enzyme-triggered release and fluorescent dye into gold nanoclusters for endomicroscopy-guided photothermal/photodynamic therapy to pancreatic ductal adenocarcinoma. Biomaterials 2017; 139:30-38. [PMID: 28582716 DOI: 10.1016/j.biomaterials.2017.05.030] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 05/18/2017] [Accepted: 05/19/2017] [Indexed: 12/16/2022]
|
13
|
Tang Y, Shao A, Cao J, Li H, Li Q, Zeng M, Liu M, Cheng Y, Zhu W. cNGR-based synergistic-targeted NIR fluorescent probe for tracing and bioimaging of pancreatic ductal adenocarcinoma. Sci China Chem 2017. [DOI: 10.1007/s11426-017-9092-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
14
|
Spira A, Yurgelun MB, Alexandrov L, Rao A, Bejar R, Polyak K, Giannakis M, Shilatifard A, Finn OJ, Dhodapkar M, Kay NE, Braggio E, Vilar E, Mazzilli SA, Rebbeck TR, Garber JE, Velculescu VE, Disis ML, Wallace DC, Lippman SM. Precancer Atlas to Drive Precision Prevention Trials. Cancer Res 2017; 77:1510-1541. [PMID: 28373404 PMCID: PMC6681830 DOI: 10.1158/0008-5472.can-16-2346] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 01/20/2017] [Accepted: 01/20/2017] [Indexed: 02/07/2023]
Abstract
Cancer development is a complex process driven by inherited and acquired molecular and cellular alterations. Prevention is the holy grail of cancer elimination, but making this a reality will take a fundamental rethinking and deep understanding of premalignant biology. In this Perspective, we propose a national concerted effort to create a Precancer Atlas (PCA), integrating multi-omics and immunity - basic tenets of the neoplastic process. The biology of neoplasia caused by germline mutations has led to paradigm-changing precision prevention efforts, including: tumor testing for mismatch repair (MMR) deficiency in Lynch syndrome establishing a new paradigm, combinatorial chemoprevention efficacy in familial adenomatous polyposis (FAP), signal of benefit from imaging-based early detection research in high-germline risk for pancreatic neoplasia, elucidating early ontogeny in BRCA1-mutation carriers leading to an international breast cancer prevention trial, and insights into the intricate germline-somatic-immunity interaction landscape. Emerging genetic and pharmacologic (metformin) disruption of mitochondrial (mt) respiration increased autophagy to prevent cancer in a Li-Fraumeni mouse model (biology reproduced in clinical pilot) and revealed profound influences of subtle changes in mt DNA background variation on obesity, aging, and cancer risk. The elaborate communication between the immune system and neoplasia includes an increasingly complex cellular microenvironment and dynamic interactions between host genetics, environmental factors, and microbes in shaping the immune response. Cancer vaccines are in early murine and clinical precancer studies, building on the recent successes of immunotherapy and HPV vaccine immune prevention. Molecular monitoring in Barrett's esophagus to avoid overdiagnosis/treatment highlights an important PCA theme. Next generation sequencing (NGS) discovered age-related clonal hematopoiesis of indeterminate potential (CHIP). Ultra-deep NGS reports over the past year have redefined the premalignant landscape remarkably identifying tiny clones in the blood of up to 95% of women in their 50s, suggesting that potentially premalignant clones are ubiquitous. Similar data from eyelid skin and peritoneal and uterine lavage fluid provide unprecedented opportunities to dissect the earliest phases of stem/progenitor clonal (and microenvironment) evolution/diversity with new single-cell and liquid biopsy technologies. Cancer mutational signatures reflect exogenous or endogenous processes imprinted over time in precursors. Accelerating the prevention of cancer will require a large-scale, longitudinal effort, leveraging diverse disciplines (from genetics, biochemistry, and immunology to mathematics, computational biology, and engineering), initiatives, technologies, and models in developing an integrated multi-omics and immunity PCA - an immense national resource to interrogate, target, and intercept events that drive oncogenesis. Cancer Res; 77(7); 1510-41. ©2017 AACR.
Collapse
Affiliation(s)
- Avrum Spira
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Bioinformatics, Boston University School of Medicine, Boston, Massachusetts
| | - Matthew B Yurgelun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ludmil Alexandrov
- Theoretical Division, Center for Nonlinear Studies, Los Alamos National Laboratory, Los Alamos, New Mexico
| | - Anjana Rao
- Division of Signaling and Gene Expression, La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Rafael Bejar
- Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ali Shilatifard
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Olivera J Finn
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Madhav Dhodapkar
- Department of Hematology and Immunology, Yale Cancer Center, New Haven, Connecticut
| | - Neil E Kay
- Department of Hematology, Mayo Clinic Hospital, Rochester, Minnesota
| | - Esteban Braggio
- Department of Hematology, Mayo Clinic Hospital, Phoenix, Arizona
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarah A Mazzilli
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Bioinformatics, Boston University School of Medicine, Boston, Massachusetts
| | - Timothy R Rebbeck
- Division of Hematology and Oncology, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Judy E Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Victor E Velculescu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
- Department of Pathology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Mary L Disis
- Department of Medicine, Center for Translational Medicine in Women's Health, University of Washington, Seattle, Washington
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Scott M Lippman
- Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, California.
| |
Collapse
|
15
|
O'Donoghue AJ, Ivry SL, Chaudhury C, Hostetter DR, Hanahan D, Craik CS. Procathepsin E is highly abundant but minimally active in pancreatic ductal adenocarcinoma tumors. Biol Chem 2016; 397:871-81. [PMID: 27149201 PMCID: PMC5712230 DOI: 10.1515/hsz-2016-0138] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 04/20/2016] [Indexed: 12/31/2022]
Abstract
The cathepsin family of lysosomal proteases is increasingly being recognized for their altered expression in cancer and role in facilitating tumor progression. The aspartyl protease cathepsin E is overexpressed in several cancers and has been investigated as a biomarker for pancreatic ductal adenocarcinoma (PDAC). Here we show that cathepsin E expression in mouse PDAC tumors is increased by more than 400-fold when compared to healthy pancreatic tissue. Cathepsin E accumulates over the course of disease progression and accounts for more than 3% of the tumor protein in mice with end-stage disease. Through immunoblot analysis we determined that only procathepsin E exists in mouse PDAC tumors and cell lines derived from these tumors. By decreasing the pH, this procathepsion E is converted to the mature form, resulting in an increase in proteolytic activity. Although active site inhibitors can bind procathepsin E, treatment of PDAC mice with the aspartyl protease inhibitor ritonavir did not decrease tumor burden. Lastly, we used multiplex substrate profiling by mass spectrometry to identify two synthetic peptides that are hydrolyzed by procathepsin E near neutral pH. This work represents a comprehensive analysis of procathepsin E in PDAC and could facilitate the development of improved biomarkers for disease detection.
Collapse
|
16
|
Ba C, Palmiere M, Ritt J, Mertz J. Dual-modality endomicroscopy with co-registered fluorescence and phase contrast. BIOMEDICAL OPTICS EXPRESS 2016; 7:3403-3411. [PMID: 27699107 PMCID: PMC5030019 DOI: 10.1364/boe.7.003403] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/08/2016] [Accepted: 08/08/2016] [Indexed: 05/09/2023]
Abstract
We describe a dual-modality laser scanning endomicroscope that provides simultaneous fluorescence contrast based on confocal laser endomicroscopy (CLE) and phase-gradient contrast based on scanning oblique back-scattering microscopy (sOBM). The probe consists of a 2.6mm-diameter micro-objective attached to a 30,000-core flexible fiber bundle. The dual contrasts are inherently co-registered, providing complementary information on labeled and un-labeled sample structure. Proof of principle demonstrations are presented with ex-vivo mouse colon tissue.
Collapse
|
17
|
Hur C, Tramontano AC, Dowling EC, Brooks GA, Jeon A, Brugge WR, Gazelle GS, Kong CY, Pandharipande PV. Early Pancreatic Ductal Adenocarcinoma Survival Is Dependent on Size: Positive Implications for Future Targeted Screening. Pancreas 2016; 45:1062-6. [PMID: 26692444 PMCID: PMC4912943 DOI: 10.1097/mpa.0000000000000587] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) has not experienced a meaningful mortality improvement for the past few decades. Successful screening is difficult to accomplish because most PDACs present late in their natural history, and current interventions have not provided significant benefit. Our goal was to identify determinants of survival for early PDAC to help inform future screening strategies. METHODS Early PDACs from the National Cancer Institute's Surveillance, Epidemiology, and End Results Program database (2000-2010) were analyzed. We stratified by size and included carcinomas in situ (Tis). Overall cancer-specific survival was calculated. A Cox proportional hazards model was developed and the significance of key covariates for survival prediction was evaluated. RESULTS A Kaplan-Meier plot demonstrated significant differences in survival by size at diagnosis; these survival benefits persisted after adjustment for key covariates in the Cox proportional hazards analysis. In addition, relatively weaker predictors of worse survival included older age, male sex, black race, nodal involvement, tumor location within the head of the pancreas, and no surgery or radiotherapy. CONCLUSIONS For early PDAC, we found tumor size to be the strongest predictor of survival, even after adjustment for other patient characteristics. Our findings suggest that early PDAC detection can have clinical benefit, which has positive implications for future screening strategies.
Collapse
Affiliation(s)
- Chin Hur
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, Massachusetts
- Gastroenterology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Angela C. Tramontano
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, Massachusetts
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Emily C. Dowling
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, Massachusetts
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Gabriel A. Brooks
- Dana Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Alvin Jeon
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, Massachusetts
- Gastroenterology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - William R. Brugge
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, Massachusetts
- Gastroenterology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - G. Scott Gazelle
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, Massachusetts
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Chung Yin Kong
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, Massachusetts
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Pari V. Pandharipande
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, Massachusetts
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
18
|
Krishna SG, Lee JH. Appraisal of needle-based confocal laser endomicroscopy in the diagnosis of pancreatic cysts. World J Gastroenterol 2016; 22:1701-10. [PMID: 26819534 PMCID: PMC4722000 DOI: 10.3748/wjg.v22.i4.1701] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Revised: 08/20/2015] [Accepted: 10/23/2015] [Indexed: 02/06/2023] Open
Abstract
Nearly 2.5% of cross-sectional imaging studies will report a finding of a cystic pancreatic lesion. Even though most of these are incidental findings, it remains very concerning for both patients and treating clinicians. Differentiating and predicting malignant transformation in pancreatic cystic lesions is clinically challenging. Current evaluation of suspicious cystic lesions includes a combination of radiologic imaging, endoscopic ultrasound (EUS) and cyst fluid analyses. Despite these attempts, precise diagnostic stratification among non-mucinous, mucinous, and malignant cystic lesions is often not possible until surgical resection. EUS-guided needle based confocal laser endomicroscopy (nCLE) for evaluation of pancreatic cysts is emerging as a powerful technique with remarkable potential. Though limited imaging data from 3 large clinical trials (INSPECT, DETECT and CONTACT) are currently the reference standard for nCLE imaging, nonetheless these have not been validated in large studies. The aim of this review article is to review the evolving role of EUS-guided nCLE in management of pancreatic cystic lesions in terms of its significance, adverse events, limitations, and implications.
Collapse
|
19
|
Bhutani MS, Koduru P, Joshi V, Saxena P, Suzuki R, Irisawa A, Yamao K. The role of endoscopic ultrasound in pancreatic cancer screening. Endosc Ultrasound 2016; 5:8-16. [PMID: 26879161 PMCID: PMC4770628 DOI: 10.4103/2303-9027.175876] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 10/13/2015] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer (PC) is a highly lethal cancer. Despite a significant advancement in cancer treatment, the mortality rate of PC is nearly identical to the incidence rates. Early detection of tumor or its precursor lesions with dysplasia may be the most effective approach to improve survival. Screening strategies should include identification of the population at high risk of developing PC, and an intense application of screening tools with adequate sensitivity to detect PC at an early curable stage. Endoscopic ultrasound (EUS) and magnetic resonance imaging (MRI) seem to be the most promising modalities for PC screening based on the data so far. EUS had an additional advantage over MRI by being able to obtain tissue sample during the same examination. Several questions remain unanswered at this time regarding the age to begin screening, frequency of screening, management of asymptomatic pancreatic lesions detected on screening, timing of resection, and extent of surgery and impact of screening on survival. Novel techniques such as needle-based confocal laser endomicroscopy (nCLE), along with biomarkers, may be helpful to identify pancreatic lesions with more aggressive malignant potential. Further studies will hopefully lead to the development of strategies combining EUS with other technological/biological advancements that will be cost-effective and have an impact on survival.
Collapse
Affiliation(s)
- Manoop S. Bhutani
- Department of Gastroenterology, Hepatology and Nutrition, MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Pramoda Koduru
- Department of Gastroenterology, Hepatology and Nutrition, MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Virendra Joshi
- Department of Gastroenterology, Ochsner Clinic Foundation, Ochsner Cancer Institute, New Orleans, Louisiana, USA
| | - Payal Saxena
- Department of Gastroenterology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Rei Suzuki
- Department of Gastroenterology and Rheumatology, Fukushima Medical University School of Medicine, Aizuwakamatsu, Fukushima, Japan
| | - Atsushi Irisawa
- Department of Gastroenterology, Aizu Medical Center, Fukushima Medical University, Aizuwakamatsu, Fukushima, Japan
| | - Kenji Yamao
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| |
Collapse
|
20
|
Bhutani MS, Koduru P, Joshi V, Karstensen JG, Saftoiu A, Vilmann P, Giovannini M. EUS-Guided Needle-Based Confocal Laser Endomicroscopy: A Novel Technique With Emerging Applications. Gastroenterol Hepatol (N Y) 2015; 11:235-240. [PMID: 27099595 PMCID: PMC4836594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Endoscopic ultrasound (EUS) has emerged as an excellent tool for imaging the gastrointestinal tract, as well as surrounding structures. EUS-guided fine-needle aspiration (EUS-FNA) has become the standard of care for the tissue sampling of a variety of masses and lymph nodes within and around the gut, providing further diagnostic and staging information. Confocal laser endomicroscopy (CLE) is a novel endoscopic method that enables imaging at a subcellular level of resolution during endoscopy, allowing up to 1000-fold magnification of tissue and providing an optical biopsy. A new procedure that has been developed in the past few years is needle-based confocal laser endomicroscopy (nCLE), which involves a mini-CLE probe that can be passed through a 1 9-gauge needle during EUS-FNA. This enables the real-time visualization of tissue at a microscopic level, with the potential to further improve the diagnostic accuracy of EUS-FNA. The device has been studied in animals as well as in humans, and the results so far have been promising. Recently, this method has also been used for the visualization of regulatory proteins and receptors in the pancreas, setting a cornerstone for nCLE in molecular imaging. The aim of this article is to review the role of EUS-guided nCLE in modern endoscopy and its implications in molecular imaging.
Collapse
Affiliation(s)
- Manoop S Bhutani
- Dr Bhutani is a professor and Dr Koduru is a research intern in the Department of Gastroenterology, Hepatology, and Nutrition at the University of Texas MD Anderson Cancer Center in Houston, Texas. Dr Joshi is an associate professor in the Department of Gastroenterology at the Ochsner Clinic Foundation in New Orleans, Louisiana. Dr Karstensen is a research fellow, Dr Saftoiu is a visiting professor, and Dr Vilmann is a professor at the University of Copenhagen Herlev Hospital in Herlev, Denmark. Dr Saftoiu is also a professor at the Research Center of Gastroenterology and Hepatology at the University of Medicine and Pharmacy of Craiova in Craiova, Romania. Dr Giovannini is a professor at the Institut Paoli-Calmettes in Marseille, France
| | - Pramoda Koduru
- Dr Bhutani is a professor and Dr Koduru is a research intern in the Department of Gastroenterology, Hepatology, and Nutrition at the University of Texas MD Anderson Cancer Center in Houston, Texas. Dr Joshi is an associate professor in the Department of Gastroenterology at the Ochsner Clinic Foundation in New Orleans, Louisiana. Dr Karstensen is a research fellow, Dr Saftoiu is a visiting professor, and Dr Vilmann is a professor at the University of Copenhagen Herlev Hospital in Herlev, Denmark. Dr Saftoiu is also a professor at the Research Center of Gastroenterology and Hepatology at the University of Medicine and Pharmacy of Craiova in Craiova, Romania. Dr Giovannini is a professor at the Institut Paoli-Calmettes in Marseille, France
| | - Virendra Joshi
- Dr Bhutani is a professor and Dr Koduru is a research intern in the Department of Gastroenterology, Hepatology, and Nutrition at the University of Texas MD Anderson Cancer Center in Houston, Texas. Dr Joshi is an associate professor in the Department of Gastroenterology at the Ochsner Clinic Foundation in New Orleans, Louisiana. Dr Karstensen is a research fellow, Dr Saftoiu is a visiting professor, and Dr Vilmann is a professor at the University of Copenhagen Herlev Hospital in Herlev, Denmark. Dr Saftoiu is also a professor at the Research Center of Gastroenterology and Hepatology at the University of Medicine and Pharmacy of Craiova in Craiova, Romania. Dr Giovannini is a professor at the Institut Paoli-Calmettes in Marseille, France
| | - John G Karstensen
- Dr Bhutani is a professor and Dr Koduru is a research intern in the Department of Gastroenterology, Hepatology, and Nutrition at the University of Texas MD Anderson Cancer Center in Houston, Texas. Dr Joshi is an associate professor in the Department of Gastroenterology at the Ochsner Clinic Foundation in New Orleans, Louisiana. Dr Karstensen is a research fellow, Dr Saftoiu is a visiting professor, and Dr Vilmann is a professor at the University of Copenhagen Herlev Hospital in Herlev, Denmark. Dr Saftoiu is also a professor at the Research Center of Gastroenterology and Hepatology at the University of Medicine and Pharmacy of Craiova in Craiova, Romania. Dr Giovannini is a professor at the Institut Paoli-Calmettes in Marseille, France
| | - Adrian Saftoiu
- Dr Bhutani is a professor and Dr Koduru is a research intern in the Department of Gastroenterology, Hepatology, and Nutrition at the University of Texas MD Anderson Cancer Center in Houston, Texas. Dr Joshi is an associate professor in the Department of Gastroenterology at the Ochsner Clinic Foundation in New Orleans, Louisiana. Dr Karstensen is a research fellow, Dr Saftoiu is a visiting professor, and Dr Vilmann is a professor at the University of Copenhagen Herlev Hospital in Herlev, Denmark. Dr Saftoiu is also a professor at the Research Center of Gastroenterology and Hepatology at the University of Medicine and Pharmacy of Craiova in Craiova, Romania. Dr Giovannini is a professor at the Institut Paoli-Calmettes in Marseille, France
| | - Peter Vilmann
- Dr Bhutani is a professor and Dr Koduru is a research intern in the Department of Gastroenterology, Hepatology, and Nutrition at the University of Texas MD Anderson Cancer Center in Houston, Texas. Dr Joshi is an associate professor in the Department of Gastroenterology at the Ochsner Clinic Foundation in New Orleans, Louisiana. Dr Karstensen is a research fellow, Dr Saftoiu is a visiting professor, and Dr Vilmann is a professor at the University of Copenhagen Herlev Hospital in Herlev, Denmark. Dr Saftoiu is also a professor at the Research Center of Gastroenterology and Hepatology at the University of Medicine and Pharmacy of Craiova in Craiova, Romania. Dr Giovannini is a professor at the Institut Paoli-Calmettes in Marseille, France
| | - Marc Giovannini
- Dr Bhutani is a professor and Dr Koduru is a research intern in the Department of Gastroenterology, Hepatology, and Nutrition at the University of Texas MD Anderson Cancer Center in Houston, Texas. Dr Joshi is an associate professor in the Department of Gastroenterology at the Ochsner Clinic Foundation in New Orleans, Louisiana. Dr Karstensen is a research fellow, Dr Saftoiu is a visiting professor, and Dr Vilmann is a professor at the University of Copenhagen Herlev Hospital in Herlev, Denmark. Dr Saftoiu is also a professor at the Research Center of Gastroenterology and Hepatology at the University of Medicine and Pharmacy of Craiova in Craiova, Romania. Dr Giovannini is a professor at the Institut Paoli-Calmettes in Marseille, France
| |
Collapse
|