1
|
Kim M, Woo HY, Kim J, Seo AN. Claudin 18.2 Expression in Gastric Tumors and Other Tumor Types With Gastric Epithelium-like Differentiation. In Vivo 2025; 39:1540-1553. [PMID: 40295026 PMCID: PMC12042001 DOI: 10.21873/invivo.13954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND/AIM Claudin 18.2 is an emerging biomarker for claudin 18.2-targeted therapy. We investigated claudin 18.2 expression in diverse tumor types. PATIENTS AND METHODS We retrospectively analyzed 67 gastric tumors (61 surgically resected and six biopsy specimens) and 73 other tumor types (69 resected and four biopsy specimens), including those from the pancreas, hepatobiliary system, lung, ovary, uterine cervix, and others. Claudin 18.2 expression and positivity (≥75% of tumor cells showing moderate to strong membranous staining) were assessed using claudin 18 immunostaining (clone 43-14A). RESULTS Claudin 18.2 positivity was found in 47.8% (32/67) of gastric tumor samples. Epstein-Barr virus-associated gastric cancer showed a higher frequency of positivity (6/7, 85.7%), although not statistically significantly (p=0.216). Among gastric tumors from patients with lymph node or distant metastasis (n=20), four (20.0%) exhibited discrepancies in claudin 18.2 positivity between the primary and its metastasis. In other tumor types, claudin 18.2 positivity was more frequent in those with gastric epithelium-like differentiation, including pancreatic tumors (2/9, 22.2%), hepatobiliary carcinoma (2/8, 25.0%), invasive mucinous lung adenocarcinoma (4/5, 80.0%), and mucinous ovarian tumor (5/5, 100.0%) than in those with other histology (p<0.001). Interestingly, pancreatic tumors, potential candidates for claudin 18.2-targeted therapy, often exhibited reduced or lack of claudin 18.2 expression in the invasive component. CONCLUSION Overall, claudin 18.2 positivity occurred primarily in a significant proportion of gastric tumors and other tumors with gastric epithelium-like differentiation. Evaluating claudin 18.2 expression in all such tumors can benefit patients by guiding targeted therapy. Additionally, claudin 18.2 immunostaining serves as a lineage marker for gastric origin or gastric-like differentiation.
Collapse
Affiliation(s)
- Moonsik Kim
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ha Young Woo
- Department of Pathology, Chung-Ang University Gwangmyeong Hospital, Gyeonggi-do, Republic of Korea
| | - Jinhee Kim
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - An Na Seo
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea;
| |
Collapse
|
2
|
Wang J, Seo JW, Kare AJ, Schneider M, Pandrala M, Tumbale SK, Raie MN, Engudar G, Zhang N, Guo Y, Zhong X, Ferreira S, Wu B, Attardi LD, Pratx G, Iagaru A, Brunsing RL, Charville GW, Park WG, Ferrara KW. Spatial transcriptomic analysis drives PET imaging of tight junction protein expression in pancreatic cancer theranostics. Nat Commun 2024; 15:10751. [PMID: 39737976 PMCID: PMC11686138 DOI: 10.1038/s41467-024-54761-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 11/19/2024] [Indexed: 01/01/2025] Open
Abstract
Molecular imaging using positron emission tomography (PET) provides sensitive detection and mapping of molecular targets. While cancer-associated fibroblasts and integrins have been proposed as targets for imaging of pancreatic ductal adenocarcinoma (PDAC), herein, spatial transcriptomics and proteomics of human surgical samples are applied to select PDAC targets. We find that selected cancer cell surface markers are spatially correlated and provide specific cancer localization, whereas the spatial correlation between cancer markers and immune-related or fibroblast markers is low. Claudin-4 expression increases ~16 fold in cancer as compared with normal pancreas, and tight junction localization confers low background for imaging in normal tissue. We develop a peptide-based molecular imaging agent targeted to claudin-4 with accumulation to ~25% injected activity per cubic centimeter (IA/cc) in metastases and ~18% IA/cc in tumors. Our work motivates a data-driven approach to selection of molecular targets.
Collapse
Affiliation(s)
- James Wang
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Jai Woong Seo
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Aris J Kare
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
- Department of Bioengineering, Stanford University, 443 Via Ortega, Stanford, CA, USA
| | - Martin Schneider
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Mallesh Pandrala
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Spencer K Tumbale
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Marina N Raie
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Gokce Engudar
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Nisi Zhang
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Yutong Guo
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Xiaoxu Zhong
- Department of Radiation Oncology, Stanford University, 857 Blake Wilbur Drive, Stanford, CA, USA
| | - Sofia Ferreira
- Department of Radiation Oncology, Stanford University, 857 Blake Wilbur Drive, Stanford, CA, USA
| | - Bo Wu
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Laura D Attardi
- Department of Radiation Oncology, Stanford University, 857 Blake Wilbur Drive, Stanford, CA, USA
- Department of Genetics, Stanford University, 291 Campus Drive, Stanford, CA, USA
| | - Guillem Pratx
- Department of Radiation Oncology, Stanford University, 857 Blake Wilbur Drive, Stanford, CA, USA
| | - Andrei Iagaru
- Nuclear Medicine and Molecular Imaging Division, Department of Radiology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Ryan L Brunsing
- Department of Radiology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Gregory W Charville
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Walter G Park
- Department of Medicine-Gastroenterology & Hepatology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Katherine W Ferrara
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 300 Pasteur Drive, Stanford, CA, USA.
| |
Collapse
|
3
|
Bogdanski AM, van Hooft JE, Boekestijn B, Bonsing BA, Wasser MNJM, Klatte DCF, van Leerdam ME. Aspects and outcomes of surveillance for individuals at high-risk of pancreatic cancer. Fam Cancer 2024; 23:323-339. [PMID: 38619782 PMCID: PMC11255004 DOI: 10.1007/s10689-024-00368-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/24/2024] [Indexed: 04/16/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related deaths and is associated with a poor prognosis. The majority of these cancers are detected at a late stage, contributing to the bad prognosis. This underscores the need for novel, enhanced early detection strategies to improve the outcomes. While population-based screening is not recommended due to the relatively low incidence of PDAC, surveillance is recommended for individuals at high risk for PDAC due to their increased incidence of the disease. However, the outcomes of pancreatic cancer surveillance in high-risk individuals are not sorted out yet. In this review, we will address the identification of individuals at high risk for PDAC, discuss the objectives and targets of surveillance, outline how surveillance programs are organized, summarize the outcomes of high-risk individuals undergoing pancreatic cancer surveillance, and conclude with a future perspective on pancreatic cancer surveillance and novel developments.
Collapse
Affiliation(s)
- Aleksander M Bogdanski
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Jeanin E van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Bas Boekestijn
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin N J M Wasser
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Derk C F Klatte
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Desai R, Huang L, Gonzalez RS, Muthuswamy SK. Oncogenic GNAS Uses PKA-Dependent and Independent Mechanisms to Induce Cell Proliferation in Human Pancreatic Ductal and Acinar Organoids. Mol Cancer Res 2024; 22:440-451. [PMID: 38319286 PMCID: PMC10906748 DOI: 10.1158/1541-7786.mcr-23-0199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 09/26/2023] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
IMPLICATIONS The study identifies an opportunity to discover a PKA-independent pathway downstream of oncogene GNAS for managing IPMN lesions and their progression to PDAC.
Collapse
Affiliation(s)
- Ridhdhi Desai
- Cancer Research Institute, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Current Address: Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Ling Huang
- Cancer Research Institute, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Current Address: Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan
| | - Raul S. Gonzalez
- Cancer Research Institute, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Senthil K. Muthuswamy
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, NIH, Bethesda, MD, MA, 02215, USA
| |
Collapse
|
5
|
Wang J, Seo JW, Kare AJ, Schneider M, Tumbale SK, Wu B, Raie MN, Pandrala M, Iagaru A, Brunsing RL, Charville GW, Park WG, Ferrara KW. Spatial transcriptomic analysis drives PET imaging of tight junction protein expression in pancreatic cancer theranostics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.07.574209. [PMID: 38249519 PMCID: PMC10798647 DOI: 10.1101/2024.01.07.574209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
We apply spatial transcriptomics and proteomics to select pancreatic cancer surface receptor targets for molecular imaging and theranostics using an approach that can be applied to many cancers. Selected cancer surfaceome epithelial markers were spatially correlated and provided specific cancer localization, whereas the spatial correlation between cancer markers and immune- cell or fibroblast markers was low. While molecular imaging of cancer-associated fibroblasts and integrins has been proposed for pancreatic cancer, our data point to the tight junction protein claudin-4 as a theranostic target. Claudin-4 expression increased ∼16 fold in cancer as compared with normal pancreas, and the tight junction localization conferred low background for imaging in normal tissue. We developed a peptide-based molecular imaging agent targeted to claudin-4 with accumulation to ∼25% injected activity per cc (IA/cc) in metastases and ∼18% IA/cc in tumors. Our work motivates a new approach for data-driven selection of molecular targets.
Collapse
|
6
|
Bararia A, Chakraborty P, Roy P, Chattopadhay BK, Das A, Chatterjee A, Sikdar N. Emerging role of non-invasive and liquid biopsy biomarkers in pancreatic cancer. World J Gastroenterol 2023; 29:2241-2260. [PMID: 37124888 PMCID: PMC10134423 DOI: 10.3748/wjg.v29.i15.2241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/02/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023] Open
Abstract
A global increase in the incidence of pancreatic cancer (PanCa) presents a major concern and health burden. The traditional tissue-based diagnostic techniques provided a major way forward for molecular diagnostics; however, they face limitations based on diagnosis-associated difficulties and concerns surrounding tissue availability in the clinical setting. Late disease development with asymptomatic behavior is a drawback in the case of existing diagnostic procedures. The capability of cell free markers in discriminating PanCa from autoimmune pancreatitis and chronic pancreatitis along with other precancerous lesions can be a boon to clinicians. Early-stage diagnosis of PanCa can be achieved only if these biomarkers specifically discriminate the non-carcinogenic disease stage from malignancy with respect to tumor stages. In this review, we comprehensively described the non-invasive disease detection approaches and why these approaches are gaining popularity for their early-stage diagnostic capability and associated clinical feasibility.
Collapse
Affiliation(s)
- Akash Bararia
- Human Genetics Unit, Indian Statistical Institute, Kolkata 700108, India
| | - Prosenjeet Chakraborty
- Department of Molecular Biosciences, SVYASA School of Yoga and Naturopathy, Bangalore 560105, India
| | - Paromita Roy
- Department of Pathology, Tata Medical Center, Kolkata 700160, India
| | | | - Amlan Das
- Department of Biochemistry, Royal Global University, Assam 781035, India
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9061, New Zealand
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun 248007, India
| | - Nilabja Sikdar
- Human Genetics Unit, Indian Statistical Institute, Kolkata 700108, India
| |
Collapse
|
7
|
Sawai H, Kiriyama Y, Kuzuya H, Fujii Y, Ueno S, Koide S, Kurimoto M, Yamao K, Matsuo Y, Morimoto M, Koide H, Kamiya A. Adenosquamous carcinoma coexisting with intraductal papillary mucinous neoplasm of the pancreas: a case report. J Med Case Rep 2023; 17:72. [PMID: 36859393 PMCID: PMC9979475 DOI: 10.1186/s13256-023-03798-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 01/31/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Adenosquamous carcinoma of the pancreas is a rare variant, with a worse prognosis than pancreatic ductal adenocarcinoma; moreover, it has characteristic clinical and histopathological features. Studies have mentioned the differentiation of intraductal papillary mucinous neoplasms into mucinous/tubular adenocarcinomas; however, their transdifferentiation into adenosquamous carcinoma remains unclear. CASE PRESENTATION An 80-year-old Japanese woman was referred to our hospital for further examination of multiple pancreatic cysts. Enhanced computed tomography after close follow-up for 6 years revealed a new nodule with poor enhancement on the pancreatic body. Distal pancreatectomy and splenectomy were performed. Histopathological examination revealed an adenosquamous carcinoma with coexisting intraductal papillary mucinous neoplasms; moreover, the intraductal papillary mucinous neoplasms lacked continuity with the adenosquamous carcinoma. Immunohistochemical analysis revealed squamous cell carcinoma and differentiation from adenocarcinoma to squamous cell carcinoma. Gene mutation analysis revealed KRASG12D and KRASG12R mutations in adenosquamous carcinoma components and intraductal papillary mucinous neoplasm lesions, respectively, with none showing the mutation of GNAS codon 201. The final histopathological diagnosis was adenosquamous carcinoma with coexisting intraductal papillary mucinous neoplasms of the pancreas. CONCLUSIONS This is the rare case of adenosquamous carcinoma with coexisting intraductal papillary mucinous neoplasms of the pancreas. To investigate the underlying transdifferentiation pathway of intraductal papillary mucinous neoplasms into this rare subtype of pancreatic cancer, we explored gene mutation differences as a clinicopathological parameter.
Collapse
Affiliation(s)
- Hirozumi Sawai
- Department of Surgery, Narita Memorial Hospital, Hanei-Honmachi 134, Toyohashi, Aichi, 4418029, Japan.
| | - Yuka Kiriyama
- Department of Pathology, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Hiromasa Kuzuya
- Department of Surgery, Narita Memorial Hospital, Hanei-Honmachi 134, Toyohashi, Aichi, 4418029, Japan
| | - Yoshiaki Fujii
- Department of Surgery, Narita Memorial Hospital, Hanei-Honmachi 134, Toyohashi, Aichi, 4418029, Japan
| | - Shuhei Ueno
- Department of Surgery, Narita Memorial Hospital, Hanei-Honmachi 134, Toyohashi, Aichi, 4418029, Japan
| | - Shuji Koide
- Department of Surgery, Narita Memorial Hospital, Hanei-Honmachi 134, Toyohashi, Aichi, 4418029, Japan
| | - Masaaki Kurimoto
- Department of Surgery, Narita Memorial Hospital, Hanei-Honmachi 134, Toyohashi, Aichi, 4418029, Japan
| | - Kenji Yamao
- Department of Gastroenterology, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi, 467-8601, Japan
| | - Mamoru Morimoto
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi, 467-8601, Japan
| | - Hajime Koide
- Department of Surgery, Narita Memorial Hospital, Hanei-Honmachi 134, Toyohashi, Aichi, 4418029, Japan
| | - Atsushi Kamiya
- Department of Surgery, Narita Memorial Hospital, Hanei-Honmachi 134, Toyohashi, Aichi, 4418029, Japan
| |
Collapse
|
8
|
He X, Fan R, Sun J, Ren Y, Zhao X, Rui W, Yuan Y, Zou D. A model for predicting degree of malignancy in patients with intraductal papillary mucinous neoplasm. Front Oncol 2023; 13:1087852. [PMID: 36761937 PMCID: PMC9902908 DOI: 10.3389/fonc.2023.1087852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/05/2023] [Indexed: 01/26/2023] Open
Abstract
Background/Objectives There is no predictive model available to address early stage malignant intraductal papillary mucinous neoplasm (IPMN) including high grade dysplasia (HGD) and pT1a (invasive component≤0.5 cm). The aim of this study was to establish an objective and sufficient model to predict the degree of malignancy in patients with IPMN, which can be easily applied in daily practice and adopted for any type of lesion. Methods A retrospective cohort study of 309 patients who underwent surgical resection for IPMN was performed. Members of the cohort were randomly allocated to the training or testing set. A detection tree model and random forest model were used for a 3-class classification to distinguish low grade dysplasia (LGD), HGD/pT1a IPMN, and invasive intraductal papillary mucinous cancer (I-IPMC) beyond pT1a. Results Of the 309 patients, 54 (17.4%) had early stage malignancy (19 HGD, 35 pT1a), 49 (15.9%) had I-IPMC beyond pT1a, and 206 (66.7%) had LGD IPMN. We proposed a 3-class classification model using a random forest algorithm, and the model had an accuracy of 99.5% with the training set, and displayed an accuracy of 96.0% with the testing set. We used SHAP for interpretation of the model and showed the top five factors (mural nodule size, main pancreatic duct diameter, CA19-9 levels, lesion edge and common bile duct dilation) were most likely to influence the 3-class classification results in terms of interpretation of the random forest model. Conclusions This predictive model will help assess an individual's risk for different stages of IPMN malignancy and may help identify patients with IPMN who require surgery.
Collapse
Affiliation(s)
- Xiangyi He
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Fan
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Sun
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanhao Ren
- School of Mathematical Sciences, Fudan University, Shanghai, China
| | - Xuesong Zhao
- Departments of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiwei Rui
- Departments of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaozong Yuan
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Yaozong Yuan, ; Duowu Zou,
| | - Duowu Zou
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Yaozong Yuan, ; Duowu Zou,
| |
Collapse
|
9
|
La Salvia A, Persano I, Parlagreco E, Audisio A, Cani M, Brizzi MP. Pancreatic adenocarcinoma and pancreatic high-grade neuroendocrine carcinoma: two sides of the moon. Med Oncol 2022; 39:168. [PMID: 35972607 DOI: 10.1007/s12032-022-01764-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
Pancreatic adenocarcinoma is the seventh leading cause of cancer death in the world and the most common type pf pancreatic cancer. Unfortunately, less than 20% of patients are surgically resectable and the great majority of cases are treated with palliative chemotherapy with unsatisfactory results. No targeted agents or personalized approaches have been validated in the last decades. On the other side, neuroendocrine neoplasms of the pancreas are generally considered indolent tumours. However, high-grade neuroendocrine carcinoma is a rare subtype of neuroendocrine neoplasm of the pancreas (accounting up to 10% of the neuroendocrine neoplasms of the pancreas), with particularly aggressive behaviour and poor prognosis. Even in this case, the treatment is represented by palliative chemotherapy with dismal results and no personalized therapies are available, so far. Notably, the quality of life of these patients is disappointingly low and the future perspectives of more personalized diagnostic and therapeutic strategies are scarce. In this review, we discuss relevant and current information on epidemiology, pathology, diagnosis, clinical presentation, treatment and ongoing clinical trials of these two entities, in order to illustrate the two sides of the moon.
Collapse
Affiliation(s)
- Anna La Salvia
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| | - Irene Persano
- Department of Oncology, San Luigi Gonzaga Hospital, Orbassano, Italy
| | - Elena Parlagreco
- Department of Oncology, San Luigi Gonzaga Hospital, Orbassano, Italy
| | | | - Massimiliano Cani
- Department of Oncology, San Luigi Gonzaga Hospital, Orbassano, Italy
| | - Maria Pia Brizzi
- Department of Oncology, San Luigi Gonzaga Hospital, Orbassano, Italy
| |
Collapse
|
10
|
Abdellatif TE, Safwat K, Elmenshawy HH, Abdalla W, Naguib SM. Endoscopic Ultrasonography is a Promising Tool for Preoperative Prediction of the Operability of Pancreatic Carcinoma. J Gastrointest Cancer 2022; 53:197-203. [PMID: 33411255 DOI: 10.1007/s12029-020-00567-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2020] [Indexed: 10/22/2022]
Abstract
This work discussed the efficiency of endoscopic ultrasonography (EUS) as a diagnostic modality for early detection of pancreatic cancer. The present study was constructed by doing imaging modalities EUS on 24 patients admitted to surgery department in the faculty of medicine with suspected pancreatic malignancy. Pancreatic cancer was found more in older ages above 55 years predominately in males coming from rural areas presenting with abdominal pain with or without jaundice. Histopathological examination revealed that ductal adenocarcinoma represented by 70.83% of cases. This study also shows significant elevation in serum CA19-9 in the presenting cases to confirm a close relation between cancer pancreas and CA19-9 as a good biochemical marker for PC. EUS was found to be able to detect masses less than 20 mm in diameter. Moreover, EUS can accurately detect the mass nature. EUS showed correct information as regards lymph node involvement and vascular invasion which affected the results of respectability of pancreatic masses. Finally, this study clearly showed that EUS is an accurate pre-operative tool in the assessment of nodal staging, vascular invasion, and respectability in patients with pancreatic adenocarcinoma.
Collapse
Affiliation(s)
| | - Khaled Safwat
- General Surgery Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | - Wael Abdalla
- General Surgery Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Sameh Mohamed Naguib
- General Surgery Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| |
Collapse
|
11
|
Chen X, Fu R, Shao Q, Chen Y, Ye Q, Li S, He X, Zhu J. Application of artificial intelligence to pancreatic adenocarcinoma. Front Oncol 2022; 12:960056. [PMID: 35936738 PMCID: PMC9353734 DOI: 10.3389/fonc.2022.960056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/24/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Pancreatic cancer (PC) is one of the deadliest cancers worldwide although substantial advancement has been made in its comprehensive treatment. The development of artificial intelligence (AI) technology has allowed its clinical applications to expand remarkably in recent years. Diverse methods and algorithms are employed by AI to extrapolate new data from clinical records to aid in the treatment of PC. In this review, we will summarize AI's use in several aspects of PC diagnosis and therapy, as well as its limits and potential future research avenues. METHODS We examine the most recent research on the use of AI in PC. The articles are categorized and examined according to the medical task of their algorithm. Two search engines, PubMed and Google Scholar, were used to screen the articles. RESULTS Overall, 66 papers published in 2001 and after were selected. Of the four medical tasks (risk assessment, diagnosis, treatment, and prognosis prediction), diagnosis was the most frequently researched, and retrospective single-center studies were the most prevalent. We found that the different medical tasks and algorithms included in the reviewed studies caused the performance of their models to vary greatly. Deep learning algorithms, on the other hand, produced excellent results in all of the subdivisions studied. CONCLUSIONS AI is a promising tool for helping PC patients and may contribute to improved patient outcomes. The integration of humans and AI in clinical medicine is still in its infancy and requires the in-depth cooperation of multidisciplinary personnel.
Collapse
Affiliation(s)
- Xi Chen
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Ruibiao Fu
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Shao
- Department of Surgical Ward 1, Ningbo Women and Children’s Hospital, Ningbo, China
| | - Yan Chen
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Qinghuang Ye
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Sheng Li
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Xiongxiong He
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Jinhui Zhu
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Jinhui Zhu,
| |
Collapse
|
12
|
Song J, Sokoll LJ, Chan DW, Zhang Z. Validation of Serum Biomarkers That Complement CA19-9 in Detecting Early Pancreatic Cancer Using Electrochemiluminescent-Based Multiplex Immunoassays. Biomedicines 2021; 9:biomedicines9121897. [PMID: 34944713 PMCID: PMC8698985 DOI: 10.3390/biomedicines9121897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy; its early detection is critical for improving prognosis. Electrochemiluminescent-based multiplex immunoassays were developed with high analytical performance. All proteins were analyzed in sera of patients diagnosed with PDAC (n = 138), benign pancreatic conditions (111), and healthy controls (70). The clinical performance of these markers was evaluated individually or in combination for their complementarity to CA19-9 in detecting early PDAC. Logistic regression modeling including sex and age as cofactors identified a two-marker panel of CA19-9 and CA-125 that significantly improved the performance of CA19-9 alone in discriminating PDAC (AUC: 0.857 vs. 0.766), as well as early stage PDAC (0.805 vs. 0.702) from intraductal papillary mucinous neoplasm (IPMN). At a fixed specificity of 80%, the panel significantly improved sensitivities (78% vs. 41% or 72% vs. 59%). A two-marker panel of HE4 and CEA significantly outperformed CA19-9 in separating IPMN from chronic pancreatitis (0.841 vs. 0.501). The biomarker panels evaluated by assays demonstrated potential complementarity to CA19-9 in detecting early PDAC, warranting additional clinical validation to determine their role in the early detection of pancreatic cancer.
Collapse
Affiliation(s)
- Jin Song
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, 419 North Caroline Street, Baltimore, MD 21231, USA; (L.J.S.); (D.W.C.)
- Correspondence: (J.S.); (Z.Z.); Tel.: +1-443-287-6363 (J.S.); +1-410-502-7871 (Z.Z.)
| | - Lori J. Sokoll
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, 419 North Caroline Street, Baltimore, MD 21231, USA; (L.J.S.); (D.W.C.)
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Daniel W. Chan
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, 419 North Caroline Street, Baltimore, MD 21231, USA; (L.J.S.); (D.W.C.)
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Zhen Zhang
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, 419 North Caroline Street, Baltimore, MD 21231, USA; (L.J.S.); (D.W.C.)
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Correspondence: (J.S.); (Z.Z.); Tel.: +1-443-287-6363 (J.S.); +1-410-502-7871 (Z.Z.)
| |
Collapse
|
13
|
Molecular aspects of pancreatic cancer: focus on reprogrammed metabolism in a nutrient-deficient environment and potential therapeutic targets. Cent Eur J Immunol 2021; 46:258-263. [PMID: 34764796 PMCID: PMC8568029 DOI: 10.5114/ceji.2021.107027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 04/08/2021] [Indexed: 11/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is still burdened with high mortality (5-year survival rate < 9%) due to late diagnosis, aggressiveness, and a lack of more effective treatment methods. Early diagnosis and new therapeutic approaches based on the reprogrammed metabolism of the tumor in a nutrient-deficient environment are expected to improve the future treatment of PDAC patients. Research results suggest that genetic and metabolic disorders may precede the onset of neoplastic changes, which should allow for earlier appropriate treatment. Glycolysis and glutaminolysis are the most investigated pathways associated with the highest aggressiveness of pancreatic tumors. Blocking of selected metabolic pathways related to the local adaptive metabolic activity of pancreatic cancer cells improving nutrient acquisition and metabolic crosstalk within the microenvironment to sustain proliferation may inhibit cancer development, increase cancer cells death, and increase sensitivity to other forms of treatment (e.g., chemotherapy). Depriving cancer cells of important nutrients (glucose, glutamine) revealed tumor “checkpoints” for the mechanisms that drive cell proliferation and metastasis formation in order to determine its accuracy for individualization of the therapeutic approach. The present review highlights selected metabolic signaling pathways and their regulators aimed at inhibiting the neoplastic process. Particular attention has been paid to the adaptive metabolism of pancreatic cancer, which promotes its development in an oxygen-deficient and nutrient-poor environment.
Collapse
|
14
|
Cortez NE, Mackenzie GG. Ketogenic Diets in Pancreatic Cancer and Associated Cachexia: Cellular Mechanisms and Clinical Perspectives. Nutrients 2021; 13:nu13093202. [PMID: 34579079 PMCID: PMC8471358 DOI: 10.3390/nu13093202] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/06/2021] [Accepted: 09/10/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive and extremely therapy-resistant cancer. It is estimated that up to 80% of PDAC patients present with cachexia, a multifactorial disorder characterized by the involuntary and ongoing wasting of skeletal muscle that affects therapeutic response and survival. During the last decade, there has been an increased interest in exploring dietary interventions to complement the treatment of PDAC and associated cachexia. Ketogenic diets (KDs) have gained attention for their anti-tumor potential. Characterized by a very low carbohydrate, moderate protein, and high fat composition, this diet mimics the metabolic changes that occur in fasting. Numerous studies report that a KD reduces tumor growth and can act as an adjuvant therapy in various cancers, including pancreatic cancer. However, research on the effect and mechanisms of action of KDs on PDAC-associated cachexia is limited. In this narrative review, we summarize the evidence of the impact of KDs in PDAC treatment and cachexia mitigation. Furthermore, we discuss key cellular mechanisms that explain KDs’ potential anti-tumor and anti-cachexia effects, focusing primarily on reprogramming of cell metabolism, epigenome, and the gut microbiome. Finally, we provide a perspective on future research needed to advance KDs into clinical use.
Collapse
|
15
|
Chung MJ, Park SW, Kim SH, Cho CM, Choi JH, Choi EK, Lee TH, Cho E, Lee JK, Song TJ, Lee JM, Son JH, Park JS, Oh CH, Park DA, Byeon JS, Lee ST, Kim HG, Chun HJ, Choi HS, Park CG, Cho JY. [Clinical and Technical Guideline for Endoscopic Ultrasound-guided Tissue Acquisition of Pancreatic Solid Tumor: Korean Society of Gastrointestinal Endoscopy]. THE KOREAN JOURNAL OF GASTROENTEROLOGY = TAEHAN SOHWAGI HAKHOE CHI 2021; 78:73-93. [PMID: 34446631 DOI: 10.4166/kjg.2021.057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022]
Abstract
Endoscopic ultrasound (EUS)-guided tissue acquisition of pancreatic solid tumor requires a strict recommendation for its proper use in clinical practice because of its technical difficulty and invasiveness. The Korean Society of Gastrointestinal Endoscopy appointed a Task Force to draft clinical practice guidelines for EUS-guided tissue acquisition of pancreatic solid tumor. The strength of recommendation and the level of evidence for each statement were graded according to the Minds Handbook for Clinical Practice Guideline Development 2014. The committee, comprising a development panel of 16 endosonographers and an expert on guideline development methodology, developed 12 evidence-based recommendations in eight categories intended to help physicians make evidence- based clinical judgments with regard to the diagnosis of pancreatic solid tumor. This clinical practice guideline discusses EUS-guided sampling in pancreatic solid tumor and makes recommendations on circumstances that warrant its use, technical issues related to maximizing the diagnostic yield (e.g., needle type, needle diameter, adequate number of needle passes, sample obtaining techniques, and methods of specimen processing), adverse events of EUS-guided tissue acquisition, and learning-related issues. This guideline was reviewed by external experts and suggests best practices recommended based on the evidence available at the time of preparation. This guideline may not be applicable for all clinical situations and should be interpreted in light of specific situations and the availability of resources. It will be revised as necessary to cover progress and changes in technology and evidence from clinical practice.
Collapse
Affiliation(s)
- Moon Jae Chung
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Se Woo Park
- Department of Internal Medicine, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwasung, Korea
| | - Seong-Hun Kim
- Department of Internal Medicine, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonbuk National University Medical School, Jeonju, Korea
| | - Chang Min Cho
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Jun-Ho Choi
- Department of Internal Medicine, Dankook University Hospital, Dankook University College of Medicine, Cheonan, Korea
| | - Eun Kwang Choi
- Department of Internal Medicine, Jeju National University Hospital, Jeju National University College of Medicine, Jeju, Korea
| | - Tae Hoon Lee
- Department of Internal Medicine, Soonchunhyang University Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Eunae Cho
- Department of Internal Medicine, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Korea
| | - Jun Kyu Lee
- Department of Internal Medicine, Dongguk University Medical Center, Dongguk University College of Medicine, Goyang, Korea
| | - Tae Jun Song
- Department of Internal Medicine, Asan Medical Center, Ulsan University College of Medicine, Seoul, Korea
| | - Jae Min Lee
- Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Jun Hyuk Son
- Department of Internal Medicine, Inje University Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - Jin Suk Park
- Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine,Incheon, Korea
| | - Chi Hyuk Oh
- Department of Internal Medicine, KyungHee University Medical Center, Kyung Hee University College of Medicine, Seoul, Korea
| | - Dong-Ah Park
- Division of Healthcare Technology Assessment Research, Office of Health Technology Assessment Research, National Evidence-based Healthcare Collaborating Agency, Seoul, Korea
| | - Jeong-Sik Byeon
- Department of Internal Medicine, Asan Medical Center, Ulsan University College of Medicine, Seoul, Korea
| | - Soo Teik Lee
- Department of Internal Medicine, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonbuk National University Medical School, Jeonju, Korea
| | - Ho Gak Kim
- Department of Internal Medicine, Daegu Catholic University Medical Center, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Hoon Jai Chun
- Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Ho Soon Choi
- Department of Internal Medicine, Hanyang University Medical Center, Hanyang University College of Medicine, Seoul, Korea
| | - Chan Guk Park
- Department of Internal Medicine, Chosun University Hospital, Chosun University College of Medicine, Gwangju, Korea
| | - Joo Young Cho
- Department of Internal Medicine, Cha University Bundang Medical Center, Cha University, Seongnam, Korea
| |
Collapse
|
16
|
Mebendazole disrupts stromal desmoplasia and tumorigenesis in two models of pancreatic cancer. Oncotarget 2021; 12:1326-1338. [PMID: 34262644 PMCID: PMC8274724 DOI: 10.18632/oncotarget.28014] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 06/14/2021] [Indexed: 12/14/2022] Open
Abstract
The five-year survival rate for metastatic pancreatic cancer is currently only 3%, which increases to 13% with local invasion only and to 39% with localized disease at diagnosis. Here we evaluated repurposed mebendazole, an approved anthelminthic drug, to determine how mebendazole might work at the different stages of pancreatic cancer formation and progression. We asked if mebendazole could prevent initiation of pancreatic intraepithelial neoplasia precursor lesions, interfere with stromal desmoplasia, or suppress tumor growth and liver metastasis. In both the Kras LSL.G12D/+; Pdx1-Cre (KC) mouse model of caerulein-induced inflammatory pancreatitis and the Kras LSL.G12D/+; Tp53 R172H/+; Pdx1-Cre (KPC) mouse model of advanced pancreatic cancer, mebendazole significantly reduced pancreas weight, dysplasia and intraepithelial neoplasia formation, compared to controls. Mebendazole significantly reduced trichrome-positive fibrotic connective tissue and α-SMA-positive activated pancreatic stellate cells that heralds fibrogenesis. In the aggressive KPC model, mebendazole significantly suppressed pancreatic tumor growth, both as an early and late intervention. Mebendazole reduced the overall incidence of pancreatic cancer and severity of liver metastasis in KPC mice. Using early models of pancreatic cancer, treatment with mebendazole resulted in less inflammation, decreased dysplasia, with the later stage model additionally showing a decreased tumor burden, less advanced tumors, and a reduction of metastasis. We conclude that mebendazole should be investigated further as a component of adjuvant therapy to slow progression and prevent metastasis, and well as for primary prevention in the highest risk patients.
Collapse
|
17
|
Chung MJ, Park SW, Kim SH, Cho CM, Choi JH, Choi EK, Lee TH, Cho E, Lee JK, Song TJ, Lee JM, Son JH, Park JS, Oh CH, Park DA, Byeon JS, Lee ST, Kim HG, Chun HJ, Choi HS, Park CG, Cho JY. Clinical and Technical Guideline for Endoscopic Ultrasound (EUS)-Guided Tissue Acquisition of Pancreatic Solid Tumor: Korean Society of Gastrointestinal Endoscopy (KSGE). Gut Liver 2021; 15:354-374. [PMID: 33767027 PMCID: PMC8039738 DOI: 10.5946/ce.2021.069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/13/2020] [Accepted: 01/15/2021] [Indexed: 12/13/2022] Open
Abstract
Endoscopic ultrasound (EUS)-guided tissue acquisition of pancreatic solid tumor requires a strict recommendation for its proper use in clinical practice because of its technical difficulty and invasiveness. The Korean Society of Gastrointestinal Endoscopy (KSGE) appointed a task force to draft clinical practice guidelines for EUS-guided tissue acquisition of pancreatic solid tumor. The strength of recommendation and the level of evidence for each statement were graded according to the Minds Handbook for Clinical Practice Guideline Development 2014. The committee, comprising a development panel of 16 endosonographers and an expert on guideline development methodology, developed 12 evidence-based recommendations in eight categories intended to help physicians make evidence-based clinical judgments with regard to the diagnosis of pancreatic solid tumor. This clinical practice guideline discusses EUS-guided sampling in pancreatic solid tumor and makes recommendations on circumstances that warrant its use, technical issues related to maximizing the diagnostic yield (e.g., needle type, needle diameter, adequate number of needle passes, sample obtaining techniques, and methods of specimen processing), adverse events of EUS-guided tissue acquisition, and learning-related issues. This guideline was reviewed by external experts and suggests best practices recommended based on the evidence available at the time of preparation. This guideline may not be applicable for all clinical situations and should be interpreted in light of specific situations and the availability of resources. It will be revised as necessary to cover progress and changes in technology and evidence from clinical practice.
Collapse
Affiliation(s)
- Moon Jae Chung
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Se Woo Park
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Hwaseong, Korea
| | - Seong-Hun Kim
- Department of Internal Medicine, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Chang Min Cho
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Jun-Ho Choi
- Division of Gastroenterology, Department of Internal Medicine, Dankook University College of Medicine, Cheonan, Korea
| | - Eun Kwang Choi
- Division of Gastroenterology, Department of Internal Medicine, Jeju National University College of Medicine, Jeju, Korea
| | - Tae Hoon Lee
- Division of Gastroenterology, Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Eunae Cho
- Division of Gastroenterology, Department of Internal Medicine, Chonnam National University College of Medicine, Gwangju, Korea
| | - Jun Kyu Lee
- Division of Gastroenterology, Department of Internal Medicine, Dongguk University College of Medicine, Goyang, Korea
| | - Tae Jun Song
- Division of Gastroenterology, Department of Internal Medicine, Ulsan University College of Medicine, Seoul, Korea
| | - Jae Min Lee
- Division of Gastroenterology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Jun Hyuk Son
- Division of Gastroenterology, Department of Internal Medicine, Inje University College of Medicine, Goyang, Korea
| | - Jin Suk Park
- Division of Gastroenterology, Department of Internal Medicine, Inha University College of Medicine, Incheon, Korea
| | - Chi Hyuk Oh
- Division of Gastroenterology, Department of Internal Medicine, Kyung Hee University College of Medicine, Seoul, Korea
| | - Dong-Ah Park
- Division of Healthcare Technology Assessment Research, Office of Health Technology Assessment Research, National Evidence-based Healthcare Collaborating Agency, Seoul, Korea
| | - Jeong-Sik Byeon
- Division of Gastroenterology, Department of Internal Medicine, Ulsan University College of Medicine, Seoul, Korea
| | - Soo Teik Lee
- Department of Internal Medicine, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Ho Gak Kim
- Division of Gastroenterology, Department of Internal Medicine, Catholic University of Daegu College of Medicine, Daegu, Korea
| | - Hoon Jai Chun
- Division of Gastroenterology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Ho Soon Choi
- Division of Gastroenterology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Chan Guk Park
- Division of Gastroenterology, Department of Internal Medicine, Chosun University College of Medicine, Korea, Gwangju, Korea
| | - Joo Young Cho
- Division of Gastroenterology, Department of Internal Medicine, Cha University College of Medicine, Seongnam, Korea
| |
Collapse
|
18
|
Chung MJ, Park SW, Kim SH, Cho CM, Choi JH, Choi EK, Lee TH, Cho E, Lee JK, Song TJ, Lee JM, Son JH, Park JS, Oh CH, Park DA, Byeon JS, Lee ST, Kim HG, Chun HJ, Choi HS, Park CG, Cho JY. Clinical and Technical Guideline for Endoscopic Ultrasound (EUS)-Guided Tissue Acquisition of Pancreatic Solid Tumor: Korean Society of Gastrointestinal Endoscopy (KSGE). Gut Liver 2021; 15:354-374. [PMID: 33767027 PMCID: PMC8129669 DOI: 10.5009/gnl20302] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/13/2020] [Accepted: 01/15/2021] [Indexed: 12/13/2022] Open
Abstract
Endoscopic ultrasound (EUS)-guided tissue acquisition of pancreatic solid tumor requires a strict recommendation for its proper use in clinical practice because of its technical difficulty and invasiveness. The Korean Society of Gastrointestinal Endoscopy (KSGE) appointed a task force to draft clinical practice guidelines for EUS-guided tissue acquisition of pancreatic solid tumor. The strength of recommendation and the level of evidence for each statement were graded according to the Minds Handbook for Clinical Practice Guideline Development 2014. The committee, comprising a development panel of 16 endosonographers and an expert on guideline development methodology, developed 12 evidence-based recommendations in eight categories intended to help physicians make evidence-based clinical judgments with regard to the diagnosis of pancreatic solid tumor. This clinical practice guideline discusses EUS-guided sampling in pancreatic solid tumor and makes recommendations on circumstances that warrant its use, technical issues related to maximizing the diagnostic yield (e.g., needle type, needle diameter, adequate number of needle passes, sample obtaining techniques, and methods of specimen processing), adverse events of EUS-guided tissue acquisition, and learning-related issues. This guideline was reviewed by external experts and suggests best practices recommended based on the evidence available at the time of preparation. This guideline may not be applicable for all clinical situations and should be interpreted in light of specific situations and the availability of resources. It will be revised as necessary to cover progress and changes in technology and evidence from clinical practice.
Collapse
Affiliation(s)
- Moon Jae Chung
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Se Woo Park
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Hwaseong, Korea
| | - Seong-Hun Kim
- Department of Internal Medicine, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Chang Min Cho
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Jun-Ho Choi
- Division of Gastroenterology, Department of Internal Medicine, Dankook University College of Medicine, Cheonan, Korea
| | - Eun Kwang Choi
- Division of Gastroenterology, Department of Internal Medicine, Jeju National University College of Medicine, Jeju, Korea
| | - Tae Hoon Lee
- Division of Gastroenterology, Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Eunae Cho
- Division of Gastroenterology, Department of Internal Medicine, Chonnam National University College of Medicine, Gwangju, Korea
| | - Jun Kyu Lee
- Division of Gastroenterology, Department of Internal Medicine, Dongguk University College of Medicine, Goyang, Korea
| | - Tae Jun Song
- Division of Gastroenterology, Department of Internal Medicine, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Min Lee
- Division of Gastroenterology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Jun Hyuk Son
- Division of Gastroenterology, Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Jin Suk Park
- Division of Gastroenterology, Department of Internal Medicine, Inha University College of Medicine, Incheon, Korea
| | - Chi Hyuk Oh
- Division of Gastroenterology, Department of Internal Medicine, Kyung Hee University College of Medicine, Seoul, Korea
| | - Dong-Ah Park
- Division of Healthcare Technology Assessment Research, Office of Health Technology Assessment Research, National Evidence-Based Healthcare Collaborating Agency, Seoul, Korea
| | - Jeong-Sik Byeon
- Division of Gastroenterology, Department of Internal Medicine, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo Teik Lee
- Department of Internal Medicine, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Ho Gak Kim
- Division of Gastroenterology, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Hoon Jai Chun
- Division of Gastroenterology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Ho Soon Choi
- Division of Gastroenterology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Chan Guk Park
- Division of Gastroenterology, Department of Internal Medicine, Chosun University College of Medicine, Gwangju, Korea
| | - Joo Young Cho
- Division of Gastroenterology, Department of Internal Medicine, Cha University College of Medicine, Seongnam, Korea
| |
Collapse
|
19
|
Usefulness of Deep Learning Analysis for the Diagnosis of Malignancy in Intraductal Papillary Mucinous Neoplasms of the Pancreas. Clin Transl Gastroenterol 2020; 10:1-8. [PMID: 31117111 PMCID: PMC6602761 DOI: 10.14309/ctg.0000000000000045] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Intraductal papillary mucinous neoplasms (IPMNs) are precursor lesions of pancreatic adenocarcinoma. Artificial intelligence (AI) is a mathematical concept whose implementation automates learning and recognizing data patterns. The aim of this study was to investigate whether AI via deep learning algorithms using endoscopic ultrasonography (EUS) images of IPMNs could predict malignancy.
Collapse
|
20
|
Hasan N, Ahuja N. The Emerging Roles of ATP-Dependent Chromatin Remodeling Complexes in Pancreatic Cancer. Cancers (Basel) 2019; 11:E1859. [PMID: 31769422 PMCID: PMC6966483 DOI: 10.3390/cancers11121859] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 02/08/2023] Open
Abstract
Pancreatic cancer is an aggressive cancer with low survival rates. Genetic and epigenetic dysregulation has been associated with the initiation and progression of pancreatic tumors. Multiple studies have pointed to the involvement of aberrant chromatin modifications in driving tumor behavior. ATP-dependent chromatin remodeling complexes regulate chromatin structure and have critical roles in stem cell maintenance, development, and cancer. Frequent mutations and chromosomal aberrations in the genes associated with subunits of the ATP-dependent chromatin remodeling complexes have been detected in different cancer types. In this review, we summarize the current literature on the genomic alterations and mechanistic studies of the ATP-dependent chromatin remodeling complexes in pancreatic cancer. Our review is focused on the four main subfamilies: SWItch/sucrose non-fermentable (SWI/SNF), imitation SWI (ISWI), chromodomain-helicase DNA-binding protein (CHD), and INOsitol-requiring mutant 80 (INO80). Finally, we discuss potential novel treatment options that use small molecules to target these complexes.
Collapse
Affiliation(s)
| | - Nita Ahuja
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06520, USA;
| |
Collapse
|
21
|
Xu Y, Liu J, Nipper M, Wang P. Ductal vs. acinar? Recent insights into identifying cell lineage of pancreatic ductal adenocarcinoma. ACTA ACUST UNITED AC 2019; 2. [PMID: 31528855 DOI: 10.21037/apc.2019.06.03] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease with a 5-year survival rate of less than 8%. To date, there are no early detection methods or effective treatments available. Many questions remain to be answered in regards to the pathogenesis of PDAC, among which, the controversy over the cell lineage of PDAC demands more attention. Ductal cells were originally thought to be the cell of origin for PDAC due to the ductal morphology of most cases of PDAC. However, recent studies have demonstrated that acinar cells are more sensitive to KRAS mutation and tend to develop to PanIN and PDAC effectively, very likely by undergoing acinar to ductal metaplasia into a transient state that contributes to PDAC initiation. There is also evidence that both ductal and acinar cells can potentially develop to PDAC when exposed to certain genetic settings and stimuli, suggesting that more scrutiny is required for the identification of the true cell lineage of individual cases of PDAC. In this work, we summarize recent findings in the identification of the cellular origin of PDAC, with the goal of advancing our knowledge on the initiation and progression of the disease. We also discuss various models and techniques for investigating early events of PDAC. Better understanding of these cellular events is crucial to identify new methods for the early diagnosis and treatment of PDAC.
Collapse
Affiliation(s)
- Yi Xu
- Department of Cell Systems and Anatomy, UT Health San Antonio, TX 78229, USA
| | - Jun Liu
- Department of Cell Systems and Anatomy, UT Health San Antonio, TX 78229, USA
| | - Michael Nipper
- Department of Cell Systems and Anatomy, UT Health San Antonio, TX 78229, USA
| | - Pei Wang
- Department of Cell Systems and Anatomy, UT Health San Antonio, TX 78229, USA
| |
Collapse
|
22
|
Hasselluhn MC, Schmidt GE, Ellenrieder V, Johnsen SA, Hessmann E. Aberrant NFATc1 signaling counteracts TGFβ-mediated growth arrest and apoptosis induction in pancreatic cancer progression. Cell Death Dis 2019; 10:446. [PMID: 31171768 PMCID: PMC6554303 DOI: 10.1038/s41419-019-1682-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 05/13/2019] [Accepted: 05/20/2019] [Indexed: 12/21/2022]
Abstract
Given its aggressive tumor biology and its exceptional therapy resistance, pancreatic ductal adenocarcinoma (PDAC) remains a major challenge in cancer medicine and is characterized by a 5-year survival rate of <8%. At the cellular level, PDAC is largely driven by the activation of signaling pathways that eventually converge in altered, tumor-promoting transcription programs. In this study, we sought to determine the interplay between transforming growth factor β (TGFβ) signaling and activation of the inflammatory transcription factor nuclear factor of activated T cells (NFATc1) in the regulation of transcriptional programs throughout PDAC progression. Genome-wide transcriptome analysis and functional studies performed in primary PDAC cells and transgenic mice linked nuclear NFATc1 expression with pro-proliferative and anti-apoptotic gene signatures. Consistently, NFATc1 depletion resulted in downregulation of target genes associated with poor PDAC outcome and delayed pancreatic carcinogenesis in vivo. In contrast to previous reports and consistent with a concept of retained tumor suppressive TGFβ activity, even in established PDAC, TGFβ treatment reduced PDAC cell proliferation and promoted apoptosis even in the presence of oncogenic NFATc1. However, combined TGFβ treatment and NFATc1 depletion resulted in a tremendous abrogation of tumor-promoting gene signatures and functions. Chromatin studies implied that TGFβ-dependent regulators compete with NFATc1 for the transcriptional control of jointly regulated target genes associated with an unfavorable PDAC prognosis. Together, our findings suggest opposing consequences of TGFβ and NFATc1 activity in the regulation of pro-tumorigenic transcription programs in PDAC and emphasize the strong context-dependency of key transcription programs in the progression of this devastating disease.
Collapse
Affiliation(s)
- Marie C Hasselluhn
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center, Goettingen, Germany
| | - Geske E Schmidt
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center, Goettingen, Germany
| | - Volker Ellenrieder
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center, Goettingen, Germany
| | - Steven A Johnsen
- Department of General, Visceral and Pediatric Surgery, University Medical Center, Goettingen, Germany
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Elisabeth Hessmann
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center, Goettingen, Germany.
| |
Collapse
|
23
|
Inflammation, Biomarkers and Immuno-Oncology Pathways in Pancreatic Cancer. J Pers Med 2019; 9:jpm9020020. [PMID: 31035449 PMCID: PMC6616860 DOI: 10.3390/jpm9020020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/16/2019] [Accepted: 04/23/2019] [Indexed: 12/27/2022] Open
Abstract
It is estimated that pancreatic cancer will be the second leading cause of cancer-related deaths globally by 2030, highlighting the ongoing lack of effective treatment options for this devastating condition. There is a lack of reliable prognostic or predictive markers in pancreatic cancer to guide management decisions, whether for systemic chemotherapy, molecularly targeted therapies, or immunotherapies. To date, the results for targeted agents and immunotherapies in unselected populations of chemo-refractory pancreatic cancer have not met expectations. The reasons for this lack of efficacy of immunotherapy in pancreatic cancer are not completely understood. The challenges in pancreatic cancer include the physical barrier created by the dense desmoplastic stroma surrounding the tumor, chemokine-mediated exclusion of T cells, relatively poorer antigenicity compared to other solid tumors, paucity of infiltrating T cells within the tumor, ultimately leading to an immunosuppressive microenvironment. A better understanding of the role of inflammation in pancreatic cancer, its tumor microenvironment and individualized patient-related features, be they molecular, clinical or histopathological, would enable a more effective tailored approach to the management of pancreatic cancer. In this review, the role of inflammation, the immune tumor microenvironment and potential immune biomarkers in pancreatic cancer are explored.
Collapse
|
24
|
Riva G, Pea A, Pilati C, Fiadone G, Lawlor RT, Scarpa A, Luchini C. Histo-molecular oncogenesis of pancreatic cancer: From precancerous lesions to invasive ductal adenocarcinoma. World J Gastrointest Oncol 2018; 10:317-327. [PMID: 30364837 PMCID: PMC6198304 DOI: 10.4251/wjgo.v10.i10.317] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/13/2018] [Accepted: 08/12/2018] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer is a lethal malignancy, whose precursor lesions are pancreatic intraepithelial neoplasm, intraductal papillary mucinous neoplasm, intraductal tubulopapillary neoplasm, and mucinous cystic neoplasm. To better understand the biology of pancreatic cancer, it is fundamental to know its precursors and to study the mechanisms of carcinogenesis. Each of these precursors displays peculiar histological features, as well as specific molecular alterations. Starting from such pre-invasive lesions, this review aims at summarizing the most important aspects of carcinogenesis of pancreatic cancer, with a specific focus on the recent advances and the future perspectives of the research on this lethal tumor type.
Collapse
Affiliation(s)
- Giulio Riva
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona 37134, Italy
| | - Antonio Pea
- Department of Surgery, University and Hospital trust of Verona, Verona 37134, Italy
| | - Camilla Pilati
- Personalized Medicine, Pharmacogenomics, Therapeutic Optimization, Paris-Descartes University, Paris 75006, France
| | - Giulia Fiadone
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona 37134, Italy
| | - Rita Teresa Lawlor
- ARC-Net Research Center, University and Hospital Trust of Verona, Verona 37134, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona 37134, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona 37134, Italy
| |
Collapse
|
25
|
Sandrasegaran K, Lin Y, Asare-Sawiri M, Taiyini T, Tann M. CT texture analysis of pancreatic cancer. Eur Radiol 2018; 29:1067-1073. [PMID: 30116961 DOI: 10.1007/s00330-018-5662-1] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/15/2018] [Accepted: 07/13/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVES We investigated the value of CT texture analysis (CTTA) in predicting prognosis of unresectable pancreatic cancer. METHODS Sixty patients with unresectable pancreatic cancers at presentation were enrolled for post-processing with CTTA using commercially available software (TexRAD Ltd, Cambridge, UK). The largest cross-section of the tumour on axial CT was chosen to draw a region-of-interest. CTTA parameters (mean value of positive pixels (MPP), kurtosis, entropy, skewness), arterial and venous invasion, metastatic disease and tumour size were correlated with overall and progression-free survivals. RESULTS The median overall and progression-free survivals of cohort were 13.3 and 7.8 months, respectively. On multivariate Cox proportional hazard regression analysis, presence of metastatic disease at presentation had the highest association with overall survival (p = 0.003-0.05) and progression-free survival (p < 0.001 to p = 0.004). MPP at medium spatial filter was significantly associated with poor overall survival (p = 0.04). On Kaplan-Meier survival analysis of CTTA parameters at medium spatial filter, MPP of more than 31.625 and kurtosis of more than 0.565 had significantly worse overall survival (p = 0.036 and 0.028, respectively). CONCLUSIONS CTTA features were significantly associated with overall survival in pancreas cancer, particularly in patients with non-metastatic, locally advanced disease. KEY POINTS • CT texture analysis is easy to perform on contrast-enhanced CT. • CT texture analysis can determine prognosis in patients with unresectable pancreas cancer. • The best predictors of poor prognosis were high kurtosis and MPP.
Collapse
Affiliation(s)
- Kumar Sandrasegaran
- Department of Radiology, Indiana University School of Medicine, 550 N. University Blvd., UH 0279, Indianapolis, IN, 46202, USA.
| | - Yuning Lin
- Department of Radiology, Indiana University School of Medicine, 550 N. University Blvd., UH 0279, Indianapolis, IN, 46202, USA.,Department of Medical Imaging, Fuzhou General Hospital, Fuzhou, China
| | - Michael Asare-Sawiri
- Department of Radiology, Indiana University School of Medicine, 550 N. University Blvd., UH 0279, Indianapolis, IN, 46202, USA.,Hope Radiation Cancer, Panama City, FL, USA
| | - Tai Taiyini
- Department of Radiology, Indiana University School of Medicine, 550 N. University Blvd., UH 0279, Indianapolis, IN, 46202, USA
| | - Mark Tann
- Department of Radiology, Indiana University School of Medicine, 550 N. University Blvd., UH 0279, Indianapolis, IN, 46202, USA
| |
Collapse
|
26
|
Knudsen ES, Balaji U, Mannakee B, Vail P, Eslinger C, Moxom C, Mansour J, Witkiewicz AK. Pancreatic cancer cell lines as patient-derived avatars: genetic characterisation and functional utility. Gut 2018; 67:508-520. [PMID: 28073890 PMCID: PMC5868284 DOI: 10.1136/gutjnl-2016-313133] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 12/05/2016] [Accepted: 12/07/2016] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma (PDAC) is a therapy recalcitrant disease with the worst survival rate of common solid tumours. Preclinical models that accurately reflect the genetic and biological diversity of PDAC will be important for delineating features of tumour biology and therapeutic vulnerabilities. DESIGN 27 primary PDAC tumours were employed for genetic analysis and development of tumour models. Tumour tissue was used for derivation of xenografts and cell lines. Exome sequencing was performed on the originating tumour and developed models. RNA sequencing, histological and functional analyses were employed to determine the relationship of the patient-derived models to clinical presentation of PDAC. RESULTS The cohort employed captured the genetic diversity of PDAC. From most cases, both cell lines and xenograft models were developed. Exome sequencing confirmed preservation of the primary tumour mutations in developed cell lines, which remained stable with extended passaging. The level of genetic conservation in the cell lines was comparable to that observed with patient-derived xenograft (PDX) models. Unlike historically established PDAC cancer cell lines, patient-derived models recapitulated the histological architecture of the primary tumour and exhibited metastatic spread similar to that observed clinically. Detailed genetic analyses of tumours and derived models revealed features of ex vivo evolution and the clonal architecture of PDAC. Functional analysis was used to elucidate therapeutic vulnerabilities of relevance to treatment of PDAC. CONCLUSIONS These data illustrate that with the appropriate methods it is possible to develop cell lines that maintain genetic features of PDAC. Such models serve as important substrates for analysing the significance of genetic variants and create a unique biorepository of annotated cell lines and xenografts that were established simultaneously from same primary tumour. These models can be used to infer genetic and empirically determined therapeutic sensitivities that would be germane to the patient.
Collapse
Affiliation(s)
- Erik S Knudsen
- University of Arizona Department of Medicine, University of Arizona, Tucson, Arizona, USA,University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA,McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Uthra Balaji
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Brian Mannakee
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA
| | - Paris Vail
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA
| | - Cody Eslinger
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Christopher Moxom
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John Mansour
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Agnieszka K Witkiewicz
- University of Arizona Department of Medicine, University of Arizona, Tucson, Arizona, USA,University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA,McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA,University of Arizona Department of Pathology, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
27
|
Muñoz AR, Chakravarthy D, Gong J, Halff GA, Ghosh R, Kumar AP. Pancreatic cancer: Current status and Challenges. CURRENT PHARMACOLOGY REPORTS 2017; 3:396-408. [PMID: 29404265 PMCID: PMC5795623 DOI: 10.1007/s40495-017-0112-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF THE REVIEW The 5-year survival rate of patients with pancreatic cancer (PanCA) has remained stagnant. Unfortunately, the incidence is almost equal to mortality rates. These facts underscore the importance of concerted efforts to understand the pathology of this disease. Deregulation of multiple signaling pathways involved in a wide variety of cellular processes including proliferation, apoptosis, invasion, and metastasis contribute not only to cancer development but also to therapeutic resistance. The purpose of this review is to summarize current understanding of etiological factors including emerging evidence on the role of infectious agents, factors associated with therapeutic resistance and therapeutic options. RECENT FINDINGS The unique aspect of PanCA is "desmoplasia", a process that involves proliferation of stromal fibroblasts and collagen deposition in and around the filtrating cancer. Recent studies have identified pancreatic stellate cells (PSCs) as a potential source of such desmoplasia. Biphasic interactions between PSCs and cancer cells, endothelial cells, and/or myeloid derived suppressor cells in the tumor microenvironment contribute to pancreatic carcinogenesis. SUMMARY We summarize limitations of current therapeutic approaches and potential strategies to overcome these limitations using natural products including botanicals as adjuvant/neo-adjuvant for effective management of PanCA.
Collapse
Affiliation(s)
- Amanda R Muñoz
- Department of Urology, The University of Texas Health Science Center, San Antonio, TX
| | | | | | - Glenn A Halff
- Department of Surgery, The University of Texas Health Science Center, San Antonio, TX
| | - Rita Ghosh
- Department of Urology, The University of Texas Health Science Center, San Antonio, TX
- Department of Molecular Medicine, The University of Texas Health Science Center, San Antonio, TX
- Department of Pharmacology, The University of Texas Health Science Center, San Antonio, TX
- UT Health San Antonio Cancer Center, The University of Texas Health Science Center, San Antonio, TX
| | - Addanki P Kumar
- Department of Urology, The University of Texas Health Science Center, San Antonio, TX
- Department of Molecular Medicine, The University of Texas Health Science Center, San Antonio, TX
- Department of Pharmacology, The University of Texas Health Science Center, San Antonio, TX
- UT Health San Antonio Cancer Center, The University of Texas Health Science Center, San Antonio, TX
- South Texas Veterans Health Care System, The University of Texas Health Science Center, San Antonio, TX
| |
Collapse
|
28
|
Ferreira RMM, Sancho R, Messal HA, Nye E, Spencer-Dene B, Stone RK, Stamp G, Rosewell I, Quaglia A, Behrens A. Duct- and Acinar-Derived Pancreatic Ductal Adenocarcinomas Show Distinct Tumor Progression and Marker Expression. Cell Rep 2017; 21:966-978. [PMID: 29069604 PMCID: PMC5668631 DOI: 10.1016/j.celrep.2017.09.093] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 08/05/2017] [Accepted: 09/27/2017] [Indexed: 12/24/2022] Open
Abstract
The cell of origin of pancreatic ductal adenocarcinoma (PDAC) has been controversial. Here, we show that identical oncogenic drivers trigger PDAC originating from both ductal and acinar cells with similar histology but with distinct pathophysiology and marker expression dependent on cell of origin. Whereas acinar-derived tumors exhibited low AGR2 expression and were preceded by pancreatic intraepithelial neoplasias (PanINs), duct-derived tumors displayed high AGR2 and developed independently of a PanIN stage via non-mucinous lesions. Using orthotopic transplantation and chimera experiments, we demonstrate that PanIN-like lesions can be induced by PDAC as bystanders in adjacent healthy tissues, explaining the co-existence of mucinous and non-mucinous lesions and highlighting the need to distinguish between true precursor PanINs and PanIN-like bystander lesions. Our results suggest AGR2 as a tool to stratify PDAC according to cell of origin, highlight that not all PanIN-like lesions are precursors of PDAC, and add an alternative progression route to the current model of PDAC development.
Collapse
Affiliation(s)
- Rute M M Ferreira
- Adult Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Rocio Sancho
- Adult Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Hendrik A Messal
- Adult Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Emma Nye
- Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Bradley Spencer-Dene
- Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Richard K Stone
- Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Gordon Stamp
- Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ian Rosewell
- Transgenic Service-Biological Research Facility, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Alberto Quaglia
- King's College Hospital/King's College London, Institute of Liver Studies, Denmark Hill, London SE5 9RS, UK
| | - Axel Behrens
- Adult Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; King's College London, Faculty of Life Sciences and Medicine, Guy's Campus, London SE1 1UL, UK.
| |
Collapse
|
29
|
Martínez de Juan F, Reolid Escribano M, Martínez Lapiedra C, Maia de Alcantara F, Caballero Soto M, Calatrava Fons A, Machado I. Pancreatic adenosquamous carcinoma and intraductal papillary mucinous neoplasm in a CDKN2A germline mutation carrier. World J Gastrointest Oncol 2017; 9:390-396. [PMID: 28979722 PMCID: PMC5605340 DOI: 10.4251/wjgo.v9.i9.390] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 04/20/2017] [Accepted: 07/14/2017] [Indexed: 02/05/2023] Open
Abstract
A 69-year-old woman from a kindred with familial atypical multiple mole melanoma and carrier of a germline mutation in CDKN2A, presented with abdominal pain caused by a solid-cystic pancreatic mass. The patient had an abdominal computed tomography three years before in which there was no evidence of pancreatic lesion. The endoscopic ultrasound guided fine needle aspiration showed adenocarcinoma with squamous component. After surgical resection the final diagnosis was adenosquamous pancreatic carcinoma (ASPC) arising in an intraductal papillar mucinous neoplasm (IPMN). Adenosquamous carcinomas are uncommon in the pancreas and have rarely been described in association with IPMNs. It has worse prognosis than the ordinary pancreatic ductal adenocarcinoma and some distinct features. We review the clinical, imaging, pathologic and molecular aspects of ASPC. Differential diagnosis with contamination, squamous metaplasia and pancreatic metastases from a distant squamous carcinoma is discussed. Besides, the case is an accelerated model of the adenoma (IPMN)-carcinoma sequence probably due to the CDKN2A germline mutation. Somatic CDKN2A mutations are common events in the early steps of sporadic pancreatic cancer, but germline mutation carriers have a significantly higher risk of pancreatic carcinoma.
Collapse
Affiliation(s)
- Fernando Martínez de Juan
- Gastroenterology and Digestive Endoscopy Department, Valencian Institute of Oncology, 46009 Valencia, Spain
| | - María Reolid Escribano
- Gastroenterology and Digestive Endoscopy Department, Valencian Institute of Oncology, 46009 Valencia, Spain
| | - Carmen Martínez Lapiedra
- Gastroenterology and Digestive Endoscopy Department, Valencian Institute of Oncology, 46009 Valencia, Spain
| | - Fernanda Maia de Alcantara
- Gastroenterology and Digestive Endoscopy Department, Valencian Institute of Oncology, 46009 Valencia, Spain
| | - María Caballero Soto
- Digestive Surgery Department, Valencian Institute of Oncology, 46009 Valencia, Spain
| | - Ana Calatrava Fons
- Pathology Department, Valencian Institute of Oncology, 46009 Valencia, Spain
| | - Isidro Machado
- Pathology Department, Valencian Institute of Oncology, 46009 Valencia, Spain
| |
Collapse
|
30
|
Lee J, Snyder ER, Liu Y, Gu X, Wang J, Flowers BM, Kim YJ, Park S, Szot GL, Hruban RH, Longacre TA, Kim SK. Reconstituting development of pancreatic intraepithelial neoplasia from primary human pancreas duct cells. Nat Commun 2017; 8:14686. [PMID: 28272465 PMCID: PMC5344977 DOI: 10.1038/ncomms14686] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/24/2017] [Indexed: 12/14/2022] Open
Abstract
Development of systems that reconstitute hallmark features of human pancreatic intraepithelial neoplasia (PanINs), the precursor to pancreatic ductal adenocarcinoma, could generate new strategies for early diagnosis and intervention. However, human cell-based PanIN models with defined mutations are unavailable. Here, we report that genetic modification of primary human pancreatic cells leads to development of lesions resembling native human PanINs. Primary human pancreas duct cells harbouring oncogenic KRAS and induced mutations in CDKN2A, SMAD4 and TP53 expand in vitro as epithelial spheres. After pancreatic transplantation, mutant clones form lesions histologically similar to native PanINs, including prominent stromal responses. Gene expression profiling reveals molecular similarities of mutant clones with native PanINs, and identifies potential PanIN biomarker candidates including Neuromedin U, a circulating peptide hormone. Prospective reconstitution of human PanIN development from primary cells provides experimental opportunities to investigate pancreas cancer development, progression and early-stage detection. Models of human pancreatic intraepithelial neoplasia (PanIN) development do not exist. Here, the authors induce oncogenic KRAS and mutations in CDKN2A, SMAD4 and TP53 in primary human pancreatic cells to generate a PanIN model that recapitulates molecular and pathologic features of native PanINs.
Collapse
Affiliation(s)
- Jonghyeob Lee
- Department of Developmental Biology, Stanford University School of Medicine, 279 Campus Drive, Beckman Center B300, Stanford, California 94305, USA
| | - Emily R Snyder
- Department of Developmental Biology, Stanford University School of Medicine, 279 Campus Drive, Beckman Center B300, Stanford, California 94305, USA
| | - Yinghua Liu
- Department of Developmental Biology, Stanford University School of Medicine, 279 Campus Drive, Beckman Center B300, Stanford, California 94305, USA
| | - Xueying Gu
- Department of Developmental Biology, Stanford University School of Medicine, 279 Campus Drive, Beckman Center B300, Stanford, California 94305, USA
| | - Jing Wang
- Department of Developmental Biology, Stanford University School of Medicine, 279 Campus Drive, Beckman Center B300, Stanford, California 94305, USA
| | - Brittany M Flowers
- Department of Developmental Biology, Stanford University School of Medicine, 279 Campus Drive, Beckman Center B300, Stanford, California 94305, USA
| | - Yoo Jung Kim
- Department of Developmental Biology, Stanford University School of Medicine, 279 Campus Drive, Beckman Center B300, Stanford, California 94305, USA
| | - Sangbin Park
- Department of Developmental Biology, Stanford University School of Medicine, 279 Campus Drive, Beckman Center B300, Stanford, California 94305, USA
| | - Gregory L Szot
- UCSF Transplantation Surgery, University of California, San Francisco, San Francisco, California 94143, USA
| | - Ralph H Hruban
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | - Teri A Longacre
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, 279 Campus Drive, Beckman Center B300, Stanford, California 94305, USA
| |
Collapse
|
31
|
Kensler TW, Spira A, Garber JE, Szabo E, Lee JJ, Dong Z, Dannenberg AJ, Hait WN, Blackburn E, Davidson NE, Foti M, Lippman SM. Transforming Cancer Prevention through Precision Medicine and Immune-oncology. Cancer Prev Res (Phila) 2016; 9:2-10. [PMID: 26744449 DOI: 10.1158/1940-6207.capr-15-0406] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We have entered a transformative period in cancer prevention (including early detection). Remarkable progress in precision medicine and immune-oncology, driven by extraordinary recent advances in genome-wide sequencing, big-data analytics, blood-based technologies, and deep understanding of the tumor immune microenvironment (TME), has provided unprecedented possibilities to study the biology of premalignancy. The pace of research and discovery in precision medicine and immunoprevention has been astonishing and includes the following clinical firsts reported in 2015: driver mutations detected in circulating cell-free DNA in patients with premalignant lesions (lung); clonal hematopoiesis shown to be a premalignant state; molecular selection in chemoprevention randomized controlled trial (RCT; oral); striking efficacy in RCT of combination chemoprevention targeting signaling pathway alterations mechanistically linked to germline mutation (duodenum); molecular markers for early detection validated for lung cancer and showing promise for pancreatic, liver, and ovarian cancer. Identification of HPV as the essential cause of a major global cancer burden, including HPV16 as the single driver of an epidemic of oropharyngeal cancer in men, provides unique opportunities for the dissemination and implementation of public health interventions. Important to immunoprevention beyond viral vaccines, genetic drivers of premalignant progression were associated with increasing immunosuppressive TME; and Kras vaccine efficacy in pancreas genetically engineered mouse (GEM) model required an inhibitory adjuvant (Treg depletion). In addition to developing new (e.g., epigenetic) TME regulators, recent mechanistic studies of repurposed drugs (aspirin, metformin, and tamoxifen) have identified potent immune activity. Just as precision medicine and immune-oncology are revolutionizing cancer therapy, these approaches are transforming cancer prevention. Here, we set out a brief agenda for the immediate future of cancer prevention research (including a "Pre-Cancer Genome Atlas" or "PCGA"), which will involve the inter-related fields of precision medicine and immunoprevention - pivotal elements of a broader domain of personalized public health.
Collapse
Affiliation(s)
- Thomas W Kensler
- University of Pittsburgh, Pittsburgh, Pennsylvania and Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | | | - Eva Szabo
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - J Jack Lee
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Minneapolis, Minnesota
| | | | - William N Hait
- Janssen Research & Development, LLC, Raritan, New Jersey
| | | | - Nancy E Davidson
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Margaret Foti
- American Association for Cancer Research, Philadelphia, Pennsylvania
| | - Scott M Lippman
- Moores Cancer Center, University of California San Diego, La Jolla, California.
| |
Collapse
|
32
|
Hackeng WM, Hruban RH, Offerhaus GJA, Brosens LAA. Surgical and molecular pathology of pancreatic neoplasms. Diagn Pathol 2016; 11:47. [PMID: 27267993 PMCID: PMC4897815 DOI: 10.1186/s13000-016-0497-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/28/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Histologic characteristics have proven to be very useful for classifying different types of tumors of the pancreas. As a result, the major tumor types in the pancreas have long been classified based on their microscopic appearance. MAIN BODY Recent advances in whole exome sequencing, gene expression profiling, and knowledge of tumorigenic pathways have deepened our understanding of the underlying biology of pancreatic neoplasia. These advances have not only confirmed the traditional histologic classification system, but also opened new doors to early diagnosis and targeted treatment. CONCLUSION This review discusses the histopathology, genetic and epigenetic alterations and potential treatment targets of the five major malignant pancreatic tumors - pancreatic ductal adenocarcinoma, pancreatic neuroendocrine tumor, solid-pseudopapillary neoplasm, acinar cell carcinoma and pancreatoblastoma.
Collapse
MESH Headings
- Biomarkers, Tumor/genetics
- Carcinoma, Acinar Cell/diagnosis
- Carcinoma, Acinar Cell/genetics
- Carcinoma, Acinar Cell/surgery
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/surgery
- Eye Diseases, Hereditary/diagnosis
- Eye Diseases, Hereditary/genetics
- Eye Diseases, Hereditary/surgery
- Humans
- Neuroendocrine Tumors/diagnosis
- Neuroendocrine Tumors/genetics
- Neuroendocrine Tumors/surgery
- Optic Nerve Diseases/diagnosis
- Optic Nerve Diseases/genetics
- Optic Nerve Diseases/surgery
- Pancreas/pathology
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/surgery
Collapse
Affiliation(s)
- Wenzel M Hackeng
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Ralph H Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - G Johan A Offerhaus
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Lodewijk A A Brosens
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
33
|
Haage P, Schwartz CA, Scharwächter C. [Ductal adenocarcinoma and unusual differential diagnosis]. Radiologe 2016; 56:325-37. [PMID: 27000276 DOI: 10.1007/s00117-016-0090-1] [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: 11/24/2022]
Abstract
Ductal pancreatic adenocarcinoma is by far the most common solid tumor of the pancreas. It has a very poor prognosis, especially in the more advanced stages which are no longer locally confined. Due to mostly unspecific symptoms, imaging is key in the diagnostic process. Because of the widespread use of imaging techniques, incidental findings are to a greater extent discovered in the pancreas, which subsequently entail further work-up. Ductal pancreatic adenocarcinoma can be mimicked by a large number of different lesions, such as anatomical variants, peripancreatic structures and tumors, rarer primary solid pancreatic tumors, cystic tumors, metastases or different variants of pancreatitis. Additionally, a number of precursor lesions can be differentiated. The correct classification is thus important as an early diagnosis of ductal pancreatic adenocarcinoma is relevant for the prognosis and because the possibly avoidable treatment is very invasive. All major imaging techniques are principally suitable for pancreatic imaging. In addition to sonography of the abdomen, usually the baseline diagnostic tool, computed tomography (CT) with its superior spatial resolution, magnetic resonance imaging (MRI) with its good soft tissue differentiation capabilities, possibly in combination with MR cholangiopancreatography (MRCP), endosonography with its extraordinary spatial resolution, conceivably with additional endoscopic retrograde CP or the option of direct biopsy and finally positron emission tomography CT (PET-CT) as a molecular imaging tool are all particularly useful modalities. The various techniques all have its advantages and disadvantages; depending on the individual situation they may need to be combined.
Collapse
Affiliation(s)
- P Haage
- Zentrum für Radiologie HELIOS Universitätsklinikum Wuppertal, Universität Witten/Herdecke, Heusnerstr. 40, 42283, Wuppertal, Deutschland.
| | - C A Schwartz
- Zentrum für Radiologie HELIOS Universitätsklinikum Wuppertal, Universität Witten/Herdecke, Heusnerstr. 40, 42283, Wuppertal, Deutschland
| | - C Scharwächter
- Zentrum für Radiologie HELIOS Universitätsklinikum Wuppertal, Universität Witten/Herdecke, Heusnerstr. 40, 42283, Wuppertal, Deutschland
| |
Collapse
|
34
|
Hernandez YG, Lucas AL. MicroRNA in pancreatic ductal adenocarcinoma and its precursor lesions. World J Gastrointest Oncol 2016; 8:18-29. [PMID: 26798434 PMCID: PMC4714143 DOI: 10.4251/wjgo.v8.i1.18] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 10/09/2015] [Accepted: 12/02/2015] [Indexed: 02/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the 4th deadliest cancer in the United States, due to its aggressive nature, late detection, and resistance to chemotherapy. The majority of PDAC develops from 3 precursor lesions, pancreatic intraepithelial lesions (PanIN), intraductual papillary mucinous neoplasm (IPMN), and mucinous cystic neoplasm. Early detection and surgical resection can increase PDAC 5-year survival rate from 6% for Stage IV to 50% for Stage I. To date, there are no reliable biomarkers that can detect PDAC. MicroRNAs (miRNA) are small noncoding RNAs (18-25 nucleotides) that regulate gene expression by affecting translation of messenger RNA (mRNA). A large body of evidence suggests that miRNAs are dysregulated in various types of cancers. MiRNA has been profiled as a potential biomarker in pancreatic tumor tissue, blood, cyst fluid, stool, and saliva. Four miRNA biomarkers (miR-21, miR-155, miR-196, and miR-210) have been consistently dysregulated in PDAC. MiR-21, miR-155, and miR-196 have also been dysregulated in IPMN and PanIN lesions suggesting their use as early biomarkers of this disease. In this review, we explore current knowledge of miRNA sampling, miRNA dysregulation in PDAC and its precursor lesions, and advances that have been made in using miRNA as a biomarker for PDAC and its precursor lesions.
Collapse
|