1
|
Oikonomou D, Bhogal RH, Mavroeidis VK. Central pancreatectomy: An uncommon but potentially optimal choice of pancreatic resection. Hepatobiliary Pancreat Dis Int 2025; 24:119-127. [PMID: 39578167 DOI: 10.1016/j.hbpd.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 11/01/2024] [Indexed: 11/24/2024]
Abstract
Benign, premalignant or low-grade malignant pancreatic tumors are increasingly diagnosed owing to the widespread uptake of cross-sectional imaging. Surgical excision is a potential treatment option for these tumors. Pancreatoduodenectomy and distal pancreatectomy are the standard resections for tumors located in the pancreatic head-neck or body-tail, respectively, and not uncommonly sacrifice a significant amount of healthy pancreatic parenchyma. Central pancreatectomy (CP) is a parenchyma-sparing procedure, initially performed by Dagradi and Serio in 1982, in a patient with pancreatic neck insulinoma. Since then, an increasing number of cases are being performed worldwide, either via open or minimally invasive surgical access. Additionally, pancreatic enucleation is reserved for tumors < 3 cm, without involvement of the main pancreatic duct. CP remains an alternative approach in selected cases, albeit in the presence of some controversies, such as its use in early pancreatic ductal adenocarcinoma or metastatic deposits to the central aspect of the pancreas from other malignancies. In recent years, clarity is lacking as regards indications for CP, and despite accumulating evidence in favor of limited resections for suitable pancreatic tumors, no evidence-based consensus guidelines are yet available. Nevertheless, it appears that appropriate patient selection is of paramount importance to maximize the advantages of preservation of endocrine and exocrine pancreatic functions as well as to mitigate the risks of higher complication rates. In this comprehensive review, we explore the role of CP in the treatment of lesions located in the neck and proximal body of the pancreas.
Collapse
Affiliation(s)
- Dimitrios Oikonomou
- Department of HPB Surgery, King's College Hospital, Denmark Hill, London, SE5 9RS, UK
| | - Ricky H Bhogal
- Department of Academic Surgery, The Royal Marsden NHS Foundation Trust, Fulham Road, Chelsea, London SW3 6JJ, UK
| | - Vasileios K Mavroeidis
- Department of Academic Surgery, The Royal Marsden NHS Foundation Trust, Fulham Road, Chelsea, London SW3 6JJ, UK; Department of HPB Surgery, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol Royal Infirmary, Upper Maudlin St, Bristol BS2 8HW, UK.
| |
Collapse
|
2
|
Shionoya K, Sofuni A, Mukai S, Tsuchiya T, Tanaka R, Tonozuka R, Yamamoto K, Nagai K, Matsunami Y, Kojima H, Minami H, Hirakawa N, Asano K, Yamaguchi Y, Hama K, Itoi T. Evaluating the Usefulness of the Blood Apolipoprotein A2 Isoform Index for Pancreatic Cancer Diagnosis. Cancers (Basel) 2025; 17:1071. [PMID: 40227633 PMCID: PMC11987948 DOI: 10.3390/cancers17071071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Early detection of pancreatic cancer using existing tumor markers is challenging, and novel biomarkers are needed. Apolipoprotein A2 (APOA2), which is not directly produced by tumors, may help detect pancreatic cancer through mechanisms distinct from carbohydrate antigen 19-9 (CA 19-9). This study aimed to evaluate the diagnostic performance of the APOA2-isoform (APOA2-i) Index in patients with pancreatic cancer. Methods: Serum levels of the APOA2-i Index and CA 19-9 were measured in 76 patients with pancreatic cancer (Stage 0, n = 5; I, n = 4; II, n = 15; III, n = 19; and IV, n = 33) and 98 patients with non-pancreatic cancer (intraductal papillary mucinous neoplasm, n = 36; chronic pancreatitis, n = 33; pancreatic neuroendocrine neoplasm, n = 8; autoimmune pancreatitis, n = 9; and others, n = 12) to evaluate diagnostic performance. Results: APOA2 showed lower accuracy for advanced (stages II-IV) pancreatic cancer compared to CA 19-9 (sensitivity, 50.7% vs. 83.6%; sensitivity, 77.6% vs. 87.9%), but it provided superior accuracy for early-stage (stages 0 and I) detection (sensitivity, 33.3% vs. 22.2%; specificity, 66.7% vs. 59.4%). Three early-stage pancreatic cancer cases negative for CA 19-9 were detected with the APOA2-i Index, demonstrating high diagnostic accuracy for early-stage pancreatic cancer when both biomarkers are combined (sensitivity, 44.4%; specificity, 46.7%). The multivariate analysis revealed pancreatic cancer to be an independent risk factor for APOA2-i Index positivity (odds ratio [OR]: 3.48, p < 0.001), CA 19-9 positivity (OR: 25.5, p < 0.001), and positivity for either marker (OR: 13.3, p < 0.001). Conclusions: The APOA2-i Index, combined with CA 19-9, may improve early-stage pancreatic cancer detection, especially in challenging cases and for high-risk patient surveillance.
Collapse
Affiliation(s)
- Kento Shionoya
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Atsushi Sofuni
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
- Department of Clinical Oncology, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Shuntaro Mukai
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Takayoshi Tsuchiya
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Reina Tanaka
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Ryosuke Tonozuka
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Kenjiro Yamamoto
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Kazumasa Nagai
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Yukitoshi Matsunami
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Hiroyuki Kojima
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Hirohito Minami
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Noriyuki Hirakawa
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Kyoko Asano
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Yuma Yamaguchi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Kazuki Hama
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Takao Itoi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| |
Collapse
|
3
|
Mohamed G, Munir M, Rai A, Gaddam S. Pancreatic Cancer: Screening and Early Detection. Gastroenterol Clin North Am 2025; 54:205-221. [PMID: 39880528 DOI: 10.1016/j.gtc.2024.09.006] [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] [Indexed: 01/31/2025]
Abstract
Pancreatic cancer, often diagnosed at advanced stages, has poor survival rates. Effective screening aims to detect the disease early, improving outcomes. Current guidelines recommend screening high-risk groups, including those with a family history or genetic predispositions, using methods like endoscopic ultrasound and MRI. The American Gastroenterological Association and other organizations advise annual surveillance for high-risk individuals, typically starting at the age of 50 or 10 years younger than the youngest affected relative. For certain genetic syndromes, such as Peutz-Jeghers syndrome or hereditary pancreatitis, screening may begin as early as the age of 35 to 40 years.
Collapse
Affiliation(s)
- Ghada Mohamed
- Department of Internal Medicine, Lahey Hospital & Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Malak Munir
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, ST, Suite 7705, Los Angeles, CA 90048, USA
| | - Amar Rai
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, ST, Suite 7705, Los Angeles, CA 90048, USA
| | - Srinivas Gaddam
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, ST, Suite 7705, Los Angeles, CA 90048, USA.
| |
Collapse
|
4
|
Yasinzai AQK, Tareen B, Tracy K, Jamil N, Khan M, Ullah H, Raza M, Khan AU, Arif D, Waheed A, Sidhwa F, Misra S, Karki NR, Karim NA, Cavalcante L, Ullah A. Pancreatic ductal adenocarcinoma: exploring clinicopathological trends and racial disparities in a comprehensive U.S. population-based study. Clin Transl Oncol 2024; 26:2618-2628. [PMID: 38615292 DOI: 10.1007/s12094-024-03484-7] [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: 01/25/2024] [Accepted: 03/25/2024] [Indexed: 04/15/2024]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy about 50% of PDAC are metastatic at presentation. In this study, we evaluated PDAC demographics, annual trend analysis, racial disparities, survival rate, and the role of different treatment modalities in localized and metastatic disease. METHODS A total of 144,824 cases of PDAC were obtained from the SEER database from 2000 to 2018. RESULTS The median age was 69 years, with a slightly higher incidence in males (52%) and 80% of all cases were white. Among cases with available data, 43% were grade III tumors and 57% were metastatic. The most common site of metastasis was the liver (15.7%). The annual incidence has increased steadily from 2000 to 2018. The overall observed (OS) 5-year survival rate was 4.4% (95% CI 4.3-4.6%), and 5 years cause-specific survival (CSS) was 5% (95% CI 5.1-5.4%). The 5-year survival with multimodal therapy (chemotherapy, surgery, and radiation) was 22% (95% CI 20.5-22.8%). 5-year CSS for the blacks was lower at 4.7% (95% CI 4.2-5.1%) compared to the whites at 5.3% (95% CI 5.1-5.4%). Multivariate analysis found male gender and black race associated with worse prognosis. Kaplan-Meier survival analysis found multimodal therapy to have the best outcomes in all three stages. CONCLUSION PDAC is an aggressive malignancy with male gender and black race are associated with a poor prognosis. Surgery with chemoradiation was associated with the best overall survival. With steadily increasing rates of PDAC, improved treatment modalities are paramount to improving survival in these patients.
Collapse
Affiliation(s)
| | - Bisma Tareen
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | | | - Nimra Jamil
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Marjan Khan
- Internal Medicine, Marshfield Medical Center, Marshfield, USA
| | - Hafeez Ullah
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Muhammad Raza
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Amin Ullah Khan
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Dauod Arif
- Department of Oncology Developmental Therapeutics, Novant Health, Charlotte, NC, USA
| | - Abdul Waheed
- Department of Surgery, San Joaquin General Hospital, French Camp, CA, 95231, USA
| | - Feroze Sidhwa
- Department of Surgery, San Joaquin General Hospital, French Camp, CA, 95231, USA
| | - Subhasis Misra
- Department of Surgery, BayCare Health System, Tampa, FL, USA
| | - Nabin Raj Karki
- Department of Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, USA
| | - Nagla Abdel Karim
- Department of Hematology-Oncology, Inova Schar Cancer Institute, University of Virginia, Fairfax, VA, 22031, USA
| | - Ludimila Cavalcante
- Department of Oncology Developmental Therapeutics, Novant Health, Charlotte, NC, USA
| | - Asad Ullah
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| |
Collapse
|
5
|
Schmidt P, Lindemeyer J, Raut P, Schütz M, Saniternik S, Jönsson J, Endepols H, Fischer T, Quaas A, Schlößer HA, Thelen M, Grüll H. Multiparametric Characterization of the DSL-6A/C1 Pancreatic Cancer Model in Rats. Cancers (Basel) 2024; 16:1535. [PMID: 38672617 PMCID: PMC11049193 DOI: 10.3390/cancers16081535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The DSL-6A/C1 murine pancreatic ductal adenocarcinoma (PDAC) tumor model was established in Lewis rats and characterized through a comprehensive multiparametric analysis to compare it to other preclinical tumor models and explore potential diagnostic and therapeutical targets. DSL-6A/C1 tumors were histologically analyzed to elucidate PDAC features. The tumor microenvironment was studied for immune cell prevalence. Multiparametric MRI and PET imaging were utilized to characterize tumors, and 68Ga-FAPI-46-targeting cancer-associated fibroblasts (CAFs), were used to validate the histological findings. The histology confirmed typical PDAC characteristics, such as malformed pancreatic ductal malignant cells and CAFs. Distinct immune landscapes were identified, revealing an increased presence of CD8+ T cells and a decreased CD4+ T cell fraction within the tumor microenvironment. PET imaging with 68Ga-FAPI tracers exhibited strong tracer uptake in tumor tissues. The MRI parameters indicated increasing intralesional necrosis over time and elevated contrast media uptake in vital tumor areas. We have demonstrated that the DSL-6A/C1 tumor model, particularly due to its high tumorigenicity, tumor size, and 68Ga-FAPI-46 sensitivity, is a suitable alternative to established small animal models for many forms of preclinical analyses and therapeutic studies of PDAC.
Collapse
Affiliation(s)
- Patrick Schmidt
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
| | - Johannes Lindemeyer
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
| | - Pranali Raut
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
| | - Markus Schütz
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
- Faculty of Mathematics and Natural Sciences, Department of Chemistry, University of Cologne, 50937 Cologne, Germany
| | - Sven Saniternik
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
- Faculty of Mathematics and Natural Sciences, Department of Chemistry, University of Cologne, 50937 Cologne, Germany
| | - Jannika Jönsson
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
| | - Heike Endepols
- Faculty of Medicine and University Hospital of Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, 50937 Cologne, Germany;
- Faculty of Medicine and University Hospital of Cologne, Department of Nuclear Medicine, University of Cologne, 50937 Cologne, Germany;
- Nuclear Chemistry, Institute of Neuroscience and Medicine (INM-5), Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Thomas Fischer
- Faculty of Medicine and University Hospital of Cologne, Department of Nuclear Medicine, University of Cologne, 50937 Cologne, Germany;
| | - Alexander Quaas
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, 50937 Cologne, Germany;
| | - Hans Anton Schlößer
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (H.A.S.); (M.T.)
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Martin Thelen
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (H.A.S.); (M.T.)
| | - Holger Grüll
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
- Faculty of Mathematics and Natural Sciences, Department of Chemistry, University of Cologne, 50937 Cologne, Germany
| |
Collapse
|
6
|
Shionoya K, Koizumi K, Masuda S, Makazu M, Kubota J, Jinushi R, Kimura K. Difficulty in the diagnosis of pancreatic cancer based on the initial CT report: A retrospective study. Medicine (Baltimore) 2024; 103:e36224. [PMID: 38335424 PMCID: PMC10860937 DOI: 10.1097/md.0000000000036224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 02/12/2024] Open
Abstract
The role of computed tomography (CT) in the initial diagnosis of pancreatic cancer (PC) is well-known. CT reports made by radiologists are important as not all patients with PC are examined by specialists; however, some cases are not identified based on CT reports. Diagnosis via imaging of PC is sometimes difficult, and the diagnostic rate of PC and other pancreatic diseases can vary across radiologists. This study aimed to examine the diagnostic rate of PC in initial CT reports and the details of cases with diagnostic difficulties. This single-centered, retrospective study collected clinical data of 198 patients with histologically diagnosed PC between January 2018 and April 2022. Out of these contrast-enhanced CT was performed in 192 cases. PC was not reported as the main diagnosis in 18 patients (9.4%; 11 men and 7 women). Among these 18 cases, intrapancreatic mass lesions were detected in 3 (1.6%), indirect findings such as bile/pancreatic duct stenosis or dilation were detected in 5 (2.6%), and no PC-related findings were found in 10 (5.2%). The specialists suspected PC in 15 of these 18 cases based on initial CT reports. 17 cases were confirmed by endoscopic ultrasound-fine needle aspiration and one by biopsy after upper gastrointestinal endoscopy. To improve accuracy of its diagnosis, it is important that specialists provide feedback to diagnostic radiologists regarding the findings they did not report. Endoscopic ultrasound-fine needle aspiration should be performed by specialists when there is clinical information which indicates pancreatic disease of any kind.
Collapse
Affiliation(s)
- Kento Shionoya
- Gastroenterology Medicine Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Kazuya Koizumi
- Gastroenterology Medicine Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Sakue Masuda
- Gastroenterology Medicine Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Makomo Makazu
- Gastroenterology Medicine Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Jun Kubota
- Gastroenterology Medicine Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Ryuhei Jinushi
- Gastroenterology Medicine Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Karen Kimura
- Gastroenterology Medicine Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| |
Collapse
|
7
|
Tripathi S, Tabari A, Mansur A, Dabbara H, Bridge CP, Daye D. From Machine Learning to Patient Outcomes: A Comprehensive Review of AI in Pancreatic Cancer. Diagnostics (Basel) 2024; 14:174. [PMID: 38248051 PMCID: PMC10814554 DOI: 10.3390/diagnostics14020174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Pancreatic cancer is a highly aggressive and difficult-to-detect cancer with a poor prognosis. Late diagnosis is common due to a lack of early symptoms, specific markers, and the challenging location of the pancreas. Imaging technologies have improved diagnosis, but there is still room for improvement in standardizing guidelines. Biopsies and histopathological analysis are challenging due to tumor heterogeneity. Artificial Intelligence (AI) revolutionizes healthcare by improving diagnosis, treatment, and patient care. AI algorithms can analyze medical images with precision, aiding in early disease detection. AI also plays a role in personalized medicine by analyzing patient data to tailor treatment plans. It streamlines administrative tasks, such as medical coding and documentation, and provides patient assistance through AI chatbots. However, challenges include data privacy, security, and ethical considerations. This review article focuses on the potential of AI in transforming pancreatic cancer care, offering improved diagnostics, personalized treatments, and operational efficiency, leading to better patient outcomes.
Collapse
Affiliation(s)
- Satvik Tripathi
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Azadeh Tabari
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Arian Mansur
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Harika Dabbara
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Christopher P. Bridge
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Dania Daye
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
8
|
Berbís MÁ, Godino FP, Rodríguez-Comas J, Nava E, García-Figueiras R, Baleato-González S, Luna A. Radiomics in CT and MR imaging of the liver and pancreas: tools with potential for clinical application. Abdom Radiol (NY) 2024; 49:322-340. [PMID: 37889265 DOI: 10.1007/s00261-023-04071-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/28/2023]
Abstract
Radiomics allows the extraction of quantitative imaging features from clinical magnetic resonance imaging (MRI) and computerized tomography (CT) studies. The advantages of radiomics have primarily been exploited in oncological applications, including better characterization and staging of oncological lesions and prediction of patient outcomes and treatment response. The potential introduction of radiomics in the clinical setting requires the establishment of a standardized radiomics pipeline and a quality assurance program. Radiomics and texture analysis of the liver have improved the differentiation of hypervascular lesions such as adenomas, focal nodular hyperplasia, and hepatocellular carcinoma (HCC) during the arterial phase, and in the pretreatment determination of HCC prognostic factors (e.g., tumor grade, microvascular invasion, Ki-67 proliferation index). Radiomics of pancreatic CT and MR images has enhanced pancreatic ductal adenocarcinoma detection and its differentiation from pancreatic neuroendocrine tumors, mass-forming chronic pancreatitis, or autoimmune pancreatitis. Radiomics can further help to better characterize incidental pancreatic cystic lesions, accurately discriminating benign from malignant intrapancreatic mucinous neoplasms. Nonetheless, despite their encouraging results and exciting potential, these tools have yet to be implemented in the clinical setting. This non-systematic review will describe the essential steps in the implementation of the radiomics and feature extraction workflow from liver and pancreas CT and MRI studies for their potential clinical application. A succinct overview of reported radiomics applications in the liver and pancreas and the challenges and limitations of their implementation in the clinical setting is also discussed, concluding with a brief exploration of the future perspectives of radiomics in the gastroenterology field.
Collapse
Affiliation(s)
- M Álvaro Berbís
- Department of Radiology, HT Médica, San Juan de Dios Hospital, 14960, Córdoba, Spain.
- Department of Radiology, HT Médica, San Juan de Dios Hospital, Av. del Brillante, 106, 14012, Córdoba, Spain.
| | | | | | - Enrique Nava
- Department of Communications Engineering, University of Málaga, 29016, Málaga, Spain
| | - Roberto García-Figueiras
- Abdominal Imaging Section, University Clinical Hospital of Santiago, 15706, Santiago de Compostela, A Coruña, Spain
| | - Sandra Baleato-González
- Abdominal Imaging Section, University Clinical Hospital of Santiago, 15706, Santiago de Compostela, A Coruña, Spain
| | - Antonio Luna
- Department of Radiology, HT Médica, Clínica las Nieves, 23007, Jaén, Spain
| |
Collapse
|
9
|
LeBlanc M, Kang J, Costa AF. Can we rely on contrast-enhanced CT to identify pancreatic ductal adenocarcinoma? A population-based study in sensitivity and factors associated with false negatives. Eur Radiol 2023; 33:7656-7664. [PMID: 37266655 DOI: 10.1007/s00330-023-09758-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/20/2023] [Accepted: 03/27/2023] [Indexed: 06/03/2023]
Abstract
OBJECTIVES To determine the sensitivity of contrast-enhanced computed tomography (CECT) in detecting pancreatic ductal adenocarcinoma (PDAC) and identify factors associated with false negatives (FNs). METHODS Patients diagnosed with PDAC in 2014-2015 were retrospectively identified by a cancer registry. CECTs performed during the diagnostic interval were retrospectively classified as true positive (TP), indeterminate, or FN. Sensitivity TP/(TP+FN) was calculated for all CECTs and the following subgroups: protocol (uniphasic vs. biphasic); tumor size (≤ 2 cm vs. > 2 cm); and resectability (potentially resectable vs. unresectable). Multivariate logistic regression was performed to assess which of the following factors were associated with FN: clinical suspicion of PDAC; size >2 cm; presence of metastases; protocol; isoattenuating tumor; and potentially resectable disease on imaging. RESULTS In total, 176 CECTs (127 uniphasic; 49 biphasic) in 154 patients (90 men, mean age 72 ± 11 years) were included. Sensitivity was 125/149 (83.9%) overall and 87/106 (82.1%) and 38/43 (88.4%) for uniphasic and biphasic protocols, respectively. Sensitivity was decreased for tumors ≤ 2 cm (45.4% vs. 90.6%), no liver metastases (78.0% vs. 95.9%), and potentially resectable disease (65.3% vs. 93.0%). Factors significantly associated with FN were clinical suspicion (OR, 0.24, 95% CI: 0.07-0.75), size>2 cm (OR, 0.10, 95% CI: 0.02-0.44), absence of liver metastases (OR, 4.94, 95% CI: 1.29-22.99), and potentially resectable disease (OR, 4.13, 95% CI: 1.07-16.65). CONCLUSIONS In our population, the overall sensitivity of CECT to detect PDAC is 83.9%; however, this is substantially lower in several scenarios, including patients with potentially resectable disease. This finding has important implications for patient outcomes and efforts to maximize CECT sensitivity should be sought. CLINICAL RELEVANCE STATEMENT The sensitivity of CECT to detect PDAC is significantly decreased in the setting of sub-2 cm tumors and potentially resectable disease. A dedicated biphasic pancreatic CECT protocol has higher sensitivity and should be applied in patients with suspected pancreatic disease. KEY POINTS • The sensitivities of contrast-enhanced CT for the detection of PDAC were 87/106 (82.1%) and 38/43 (88.4%) for uniphasic and biphasic protocols, respectively. • The sensitivity of contrast-enhanced CT was decreased for small tumors ≤ 2 cm (45.4% vs. 90.6%), if there were no liver metastases (78.0% vs. 95.9%), and with potentially resectable disease (65.3% vs. 93.0%). • Absence of liver metastases (OR, 4.94, 95% CI: 1.29-22.99) and potentially resectable disease (OR, 4.13, 95% CI: 1.07-16.65) were associated with a false--negative (FN) CT result; suspicion of malignancy on the imaging requisition (OR, 0.24, 95% CI: 0.07-0.75) and size > 2 cm (OR, 0.10, 95% CI: 0.02-0.44) were negatively associated with FN.
Collapse
Affiliation(s)
- Max LeBlanc
- Department of Diagnostic Radiology, Queen Elizabeth II Health Sciences Centre and Dalhousie University, Victoria General Building, 3rd floor, 1276 South Park Street, Halifax, Nova Scotia, B3H 2Y9, Canada
| | - Jessie Kang
- Department of Diagnostic Radiology, Queen Elizabeth II Health Sciences Centre and Dalhousie University, Victoria General Building, 3rd floor, 1276 South Park Street, Halifax, Nova Scotia, B3H 2Y9, Canada
| | - Andreu F Costa
- Department of Diagnostic Radiology, Queen Elizabeth II Health Sciences Centre and Dalhousie University, Victoria General Building, 3rd floor, 1276 South Park Street, Halifax, Nova Scotia, B3H 2Y9, Canada.
| |
Collapse
|
10
|
Chu LC, Ahmed T, Blanco A, Javed A, Weisberg EM, Kawamoto S, Hruban RH, Kinzler KW, Vogelstein B, Fishman EK. Radiologists' Expectations of Artificial Intelligence in Pancreatic Cancer Imaging: How Good Is Good Enough? J Comput Assist Tomogr 2023; 47:845-849. [PMID: 37948357 PMCID: PMC10823576 DOI: 10.1097/rct.0000000000001503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
BACKGROUND Existing (artificial intelligence [AI]) tools in radiology are modeled without necessarily considering the expectations and experience of the end user-the radiologist. The literature is scarce on the tangible parameters that AI capabilities need to meet for radiologists to consider them useful tools. OBJECTIVE The purpose of this study is to explore radiologists' attitudes toward AI tools in pancreatic cancer imaging and to quantitatively assess their expectations of these tools. METHODS A link to the survey was posted on the www.ctisus.com website, advertised in the www.ctisus.com email newsletter, and publicized on LinkedIn, Facebook, and Twitter accounts. This survey asked participants about their demographics, practice, and current attitudes toward AI. They were also asked about their expectations of what constitutes a clinically useful AI tool. The survey consisted of 17 questions, which included 9 multiple choice questions, 2 Likert scale questions, 4 binary (yes/no) questions, 1 rank order question, and 1 free text question. RESULTS A total of 161 respondents completed the survey, yielding a response rate of 46.3% of the total 348 clicks on the survey link. The minimum acceptable sensitivity of an AI program for the detection of pancreatic cancer chosen by most respondents was either 90% or 95% at a specificity of 95%. The minimum size of pancreatic cancer that most respondents would find an AI useful at detecting was 5 mm. Respondents preferred AI tools that demonstrated greater sensitivity over those with greater specificity. Over half of respondents anticipated incorporating AI tools into their clinical practice within the next 5 years. CONCLUSION Radiologists are open to the idea of integrating AI-based tools and have high expectations regarding the performance of these tools. Consideration of radiologists' input is important to contextualize expectations and optimize clinical adoption of existing and future AI tools.
Collapse
Affiliation(s)
- Linda C. Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, Maryland
| | - Taha Ahmed
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, Maryland
| | - Alejandra Blanco
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, Maryland
| | - Ammar Javed
- Department of Surgery, New York University Grossman School of Medicine, New York, NY
| | - Edmund M. Weisberg
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, Maryland
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, Maryland
| | - Ralph H. Hruban
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kenneth W. Kinzler
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Bert Vogelstein
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, Maryland
| |
Collapse
|
11
|
Bojesen AB, Mortensen FV, Kirkegård J. Real-Time Identification of Pancreatic Cancer Cases Using Artificial Intelligence Developed on Danish Nationwide Registry Data. JCO Clin Cancer Inform 2023; 7:e2300084. [PMID: 37812754 DOI: 10.1200/cci.23.00084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/18/2023] [Accepted: 08/29/2023] [Indexed: 10/11/2023] Open
Abstract
PURPOSE Pancreatic cancer is expected to be the second leading cause of cancer-related deaths worldwide within few years. Most patients are not diagnosed in time for curative-intent treatment. Accelerating the time of diagnosis is a key component of reducing pancreatic cancer mortality. We developed and tested a dynamic algorithm aiming at proactively identifying patients with a substantially elevated risk of having undiagnosed pancreatic cancer. METHODS Machine learning methodology was applied to a live stream of nationwide Danish registry data. A hybrid case-control and prospective cohort design relying on incidence density sampling was used. Three models with minimal tuning were tested. All performance evaluation metrics were based on out-of-sample, out-of-time data in a monthly walk-forward strategy to avoid any temporal biases or inflation of performance metrics. Outcome was a diagnosis of pancreatic cancer. RESULTS Subgroups identified had a 10.1% risk of being diagnosed with pancreatic cancer within 1 year, corresponding to a number needed to screen of 9.9. When considering competing, potentially computed tomography-detectable GI cancers, this number is reduced to 5.7. The time of diagnosis can be accelerated by up to 142 days. CONCLUSION Currently available nationwide live data and computational resources are sufficient for real-time identification of individuals with at least 10.1% risk of having undiagnosed pancreatic cancer and 17.7% risk of any GI cancer in the Danish population. For prospective identification of high-risk patients, the area under the curve is not a useful indication of the positive predictive values achieved. Viable design solutions are demonstrated, which address the main shortfalls of the existing cancer prediction efforts in relation to temporal biases, leaks, and performance metric inflation. Efficacy evaluations with resection rates and mortality as end points are needed.
Collapse
Affiliation(s)
- Anders Bo Bojesen
- Department of Surgery, HPB Section, Aarhus University Hospital, Aarhus, Denmark
| | - Frank Viborg Mortensen
- Department of Surgery, HPB Section, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jakob Kirkegård
- Department of Surgery, HPB Section, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
12
|
Viriyasaranon T, Chun JW, Koh YH, Cho JH, Jung MK, Kim SH, Kim HJ, Lee WJ, Choi JH, Woo SM. Annotation-Efficient Deep Learning Model for Pancreatic Cancer Diagnosis and Classification Using CT Images: A Retrospective Diagnostic Study. Cancers (Basel) 2023; 15:3392. [PMID: 37444502 DOI: 10.3390/cancers15133392] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of this study was to develop a novel deep learning (DL) model without requiring large-annotated training datasets for detecting pancreatic cancer (PC) using computed tomography (CT) images. This retrospective diagnostic study was conducted using CT images collected from 2004 and 2019 from 4287 patients diagnosed with PC. We proposed a self-supervised learning algorithm (pseudo-lesion segmentation (PS)) for PC classification, which was trained with and without PS and validated on randomly divided training and validation sets. We further performed cross-racial external validation using open-access CT images from 361 patients. For internal validation, the accuracy and sensitivity for PC classification were 94.3% (92.8-95.4%) and 92.5% (90.0-94.4%), and 95.7% (94.5-96.7%) and 99.3 (98.4-99.7%) for the convolutional neural network (CNN) and transformer-based DL models (both with PS), respectively. Implementing PS on a small-sized training dataset (randomly sampled 10%) increased accuracy by 20.5% and sensitivity by 37.0%. For external validation, the accuracy and sensitivity were 82.5% (78.3-86.1%) and 81.7% (77.3-85.4%) and 87.8% (84.0-90.8%) and 86.5% (82.3-89.8%) for the CNN and transformer-based DL models (both with PS), respectively. PS self-supervised learning can increase DL-based PC classification performance, reliability, and robustness of the model for unseen, and even small, datasets. The proposed DL model is potentially useful for PC diagnosis.
Collapse
Affiliation(s)
- Thanaporn Viriyasaranon
- Graduate Program in System Health Science and Engineering, Division of Mechanical and Biomedical Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jung Won Chun
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic of Korea
| | - Young Hwan Koh
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic of Korea
| | - Jae Hee Cho
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Min Kyu Jung
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu 41944, Republic of 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 54907, Republic of Korea
| | - Hyo Jung Kim
- Department of Gastroenterology, Korea University Guro Hospital, Seoul 10408, Republic of Korea
| | - Woo Jin Lee
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic of Korea
| | - Jang-Hwan Choi
- Graduate Program in System Health Science and Engineering, Division of Mechanical and Biomedical Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Sang Myung Woo
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic of Korea
| |
Collapse
|
13
|
Badheeb M, Abdelrahim A, Esmail A, Umoru G, Abboud K, Al-Najjar E, Rasheed G, Alkhulaifawi M, Abudayyeh A, Abdelrahim M. Pancreatic Tumorigenesis: Precursors, Genetic Risk Factors and Screening. Curr Oncol 2022; 29:8693-8719. [PMID: 36421339 PMCID: PMC9689647 DOI: 10.3390/curroncol29110686] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022] Open
Abstract
Pancreatic cancer (PC) is a highly malignant and aggressive tumor. Despite medical advancement, the silent nature of PC results in only 20% of all cases considered resectable at the time of diagnosis. It is projected to become the second leading cause in 2030. Most pancreatic cancer cases are diagnosed in the advanced stages. Such cases are typically unresectable and are associated with a 5-year survival of less than 10%. Although there is no guideline consensus regarding recommendations for screening for pancreatic cancer, early detection has been associated with better outcomes. In addition to continued utilization of imaging and conventional tumor markers, clinicians should be aware of novel testing modalities that may be effective for early detection of pancreatic cancer in individuals with high-risk factors. The pathogenesis of PC is not well understood; however, various modifiable and non-modifiable factors have been implicated in pancreatic oncogenesis. PC detection in the earlier stages is associated with better outcomes; nevertheless, most oncological societies do not recommend universal screening as it may result in a high false-positive rate. Therefore, targeted screening for high-risk individuals represents a reasonable option. In this review, we aimed to summarize the pathogenesis, genetic risk factors, high-risk population, and screening modalities for PC.
Collapse
Affiliation(s)
- Mohamed Badheeb
- Internal Medicine Department, College of Medicine, Hadhramout University, Mukalla 50512, Yemen
| | | | - Abdullah Esmail
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX 77030, USA
- Correspondence: (A.E.); (M.A.)
| | - Godsfavour Umoru
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Karen Abboud
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Ebtesam Al-Najjar
- Faculty of Medicine and Health Sciences, University of Science and Technology, Sana’a 15201, Yemen
| | - Ghaith Rasheed
- Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
| | | | - Ala Abudayyeh
- Section of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maen Abdelrahim
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX 77030, USA
- Weill Cornell Medical College, New York, NY 14853, USA
- Cockrell Center for Advanced Therapeutic Phase I Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Correspondence: (A.E.); (M.A.)
| |
Collapse
|
14
|
|
15
|
Yan X, Lv K, Xiao M, Tan L, Gui Y, Zhang J, Chen X, Jia W, Li J. The diagnostic performance of contrast-enhanced ultrasound versus contrast-enhanced computed tomography for pancreatic carcinoma: a systematic review and meta-analysis. Transl Cancer Res 2022; 11:3645-3656. [PMID: 36388042 PMCID: PMC9641093 DOI: 10.21037/tcr-22-601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 08/09/2022] [Indexed: 10/11/2023]
Abstract
BACKGROUND Pancreatic carcinoma is a highly fatal disease, and early diagnosis is of vital importance. This meta-analysis aimed to compare the diagnostic performances of contrast-enhanced ultrasonography (CEUS) against contrast-enhanced computed tomography (CECT) for pancreatic carcinoma, using pathological results or alternative imaging modality as the gold standard. METHODS A thorough search was conducted in PubMed, EMBASE, Web of Science and Cochrane Library databases. Two investigators selected the studies and extracted the data independently. A bivariate mixed-effects regression model was used to calculate the pooled data. Subgroup analysis and meta-regression were performed to explore the causes of heterogeneity. RESULTS A total of 1,227 records were identified, of which 7 articles with 588 patients were assessed for eligibility. The overall sensitivity, specificity of CEUS and CECT with their 95% confidential intervals (95% CI) were 0.91 (0.85-0.94) and 0.88 (0.81-0.92), 0.83 (0.70-0.91) and 0.87 (0.73-0.94), respectively. The area under curve (AUC) of CEUS and CECT were 0.94 and 0.93. Subgroup analysis showed CEUS may be good at diagnosing lesions with diameters less than 2 cm. Tumor features, region and study type were the main causes of heterogeneity. CONCLUSIONS CEUS has a satisfying diagnostic performance for pancreatic carcinoma and it has high sensitivity for small pancreatic carcinomas (≤2 cm); besides, it performs well in discriminating pancreatic cancer from chronic pancreatitis. Therefore, CEUS can be a useful supplement to CECT.
Collapse
Affiliation(s)
- Xiaoyi Yan
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ke Lv
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Mengsu Xiao
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Tan
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Gui
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Zhang
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xueqi Chen
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wanying Jia
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jinglin Li
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
16
|
Luo K, Wang X, Zhang X, Liu Z, Huang S, Li R. The Value of Circulating Tumor Cells in the Prognosis and Treatment of Pancreatic Cancer. Front Oncol 2022; 12:933645. [PMID: 35860591 PMCID: PMC9293050 DOI: 10.3389/fonc.2022.933645] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/31/2022] [Indexed: 12/21/2022] Open
Abstract
In the past few decades, tumor diagnosis and treatment theory have developed in a variety of directions. The number of people dying from pancreatic cancer increases while the mortality rate of other common tumors decreases. Traditional imaging methods show the boundaries of pancreatic tumor, but they are not sufficient to judge early micrometastasis. Although carcinoembryonic antigen (CEA) and carbohydrate antigen19-9 (CA19-9) have the obvious advantages of simplicity and minimal invasiveness, these biomarkers obviously lack sensitivity and specificity. Circulating tumor cells (CTCs) have attracted attention as a non-invasive, dynamic, and real-time liquid biopsy technique for analyzing tumor characteristics. With the continuous development of new CTCs enrichment technologies, substantial progress has been made in the basic research of CTCs clinical application prospects. In many metastatic cancers, CTCs have been studied as an independent prognostic factor. This article reviews the research progress of CTCs in the treatment and prognosis of pancreatic cancer.
Collapse
|
17
|
She YM, Ge N. The value of endoscopic ultrasonography for differential diagnosis in obstructive jaundice of the distal common bile duct. Expert Rev Gastroenterol Hepatol 2022; 16:653-664. [PMID: 35793397 DOI: 10.1080/17474124.2022.2098111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Obstructive jaundice is a common clinical disease of great significance; however, diagnosing it according to etiology, especially in patients with distal obstructive jaundice is difficult. The development of endoscopic ultrasonography has improved diagnostic methods. Endoscopic ultrasonography not only improves the accuracy of conventional endoscopic ultrasound technology in etiological diagnosis, but also offers several special endoscopic ultrasound technologies for diagnosing distal obstructive jaundice of the common bile duct. What's more, endoscopic ultrasonography can be used to treat distal obstructive jaundice of common bile duct. AREAS COVERED This review discusses the diagnostic value and applications of endoscopic ultrasonography for obstructive jaundice of the distal common bile duct. EXPERT OPINION This article summarizes the value of endoscopic ultrasonography in the etiological diagnosis, relevant treatment applications of distal obstructive jaundice and the limitations of endoscopic ultrasonography in some etiologies due to the lack of clear comparison with other imaging methods. We also provide new data for the future research direction of endoscopic ultrasonography in distal obstructive jaundice.
Collapse
Affiliation(s)
- Yu Mo She
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Nan Ge
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
18
|
Mazza S, Elvo B, Conti CB, Drago A, Verga MC, Soro S, De Silvestri A, Cereatti F, Grassia R. Endoscopic ultrasound diagnostic gain over computed tomography and magnetic resonance cholang iopancreatography in defining etiology of idiopathic acute pancreatitis. World J Gastrointest Endosc 2022;14:376-386. [DOI: 10.4253/wjge.v14.i6.376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/23/2022] [Accepted: 05/23/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND About 10%-30% of acute pancreatitis remain idiopathic (IAP) even after clinical and imaging tests, including abdominal ultrasound (US), contrast-enhanced computed tomography (CECT) and magnetic resonance cholangiopancreatography (MRCP). This is a relevant issue, as up to 20% of patients with IAP have recurrent episodes and 26% of them develop chronic pancreatitis. Few data are available on the role of EUS in clarifying the etiology of IAP after failure of one or more cross-sectional techniques.
AIM To evaluate the diagnostic gain after failure of one or more previous cross-sectional exams.
METHODS We retrospectively collected data about consecutive patients with AP and at least one negative test between US, CECT and MRCP, who underwent linear EUS between January 2017 and December 2020. We investigated the EUS diagnostic yield and the EUS diagnostic gain over different combinations of these cross-sectional imaging techniques for the etiologic diagnosis of AP. Types and frequency of EUS diagnosis were also analyzed, and EUS diagnosis was compared with the clinical parameters. After EUS, patients were followed-up for a median of 31.5 mo to detect cases of pancreatitis recurrence.
RESULTS We enrolled 81 patients (63% males, mean age 61 ± 18, 23% with previous cholecystectomy, 17% with recurrent pancreatitis). Overall EUS diagnostic yield for AP etiological diagnosis was 79% (20% lithiasis, 31% acute on chronic pancreatitis, 14% pancreatic solid or cystic lesions, 5% pancreas divisum, 5% autoimmune pancreatitis, 5% ductal abnormalities), while 21% remained idiopathic. US, CECT and MRCP, taken alone or in combination, led to AP etiological diagnosis in 16 (20%) patients; among the remaining 65 patients, 49 (75%) obtained a diagnosis at EUS, with an overall EUS diagnostic gain of 61%. Sixty-eight patients had negative US; among them, EUS allowed etiological diagnosis in 59 (87%). Sixty-three patients had a negative CECT; among them, 47 (74%) obtained diagnosis with EUS. Twenty-four had a negative MRCP; among them, 20 (83%) had EUS diagnosis. Twenty-one had negative CT + MRCP, of which 17 (81%) had EUS diagnosis, with a EUS diagnostic gain of 63%. Patients with biliary etiology and without previous cholecystectomy had higher median values of alanine aminotransferase (154 vs 25, P = 0.010), aspartate aminotransferase (95 vs 29, P = 0.018), direct bilirubin (1.2 vs 0.6, P = 0.015), gamma-glutamyl transpeptidase (180 vs 48, P = 0.006) and alkaline phosphatase (150 vs 72, P = 0.015) Chronic pancreatitis diagnosis was more frequent in patients with recurrent pancreatitis at baseline (82% vs 21%, P < 0.001). During the follow-up, AP recurred in 3 patients, one of which remained idiopathic.
CONCLUSION EUS is a good test to define AP etiology. It showed a 63% diagnostic gain over CECT + MRCP. In suitable patients, EUS should always be performed in cases of IAP. Further prospective studies are needed.
Collapse
Affiliation(s)
- Stefano Mazza
- Gastroenterology and Digestive Endoscopy Unit, ASST Cremona, Cremona 26100, Italy
| | - Biagio Elvo
- Gastroenterology and Endoscopy Unit, Federico II University, Napoli 80131, Italy
| | | | - Andrea Drago
- Gastroenterology and Digestive Endoscopy Unit, ASST Cremona, Cremona 26100, Italy
| | - Maria Chiara Verga
- Gastroenterology and Digestive Endoscopy Unit, ASST Cremona, Cremona 26100, Italy
| | - Sara Soro
- Gastroenterology and Digestive Endoscopy Unit, ASST Cremona, Cremona 26100, Italy
| | - Annalisa De Silvestri
- Biometry and Clinical Epidemiology, Scientific Direction, IRCCS San Matteo Hospital Foundation, Pavia 27100, Italy
| | - Fabrizio Cereatti
- Department of Gastroenterology and Digestive Endoscopy, Castelli Hospital, Ariccia (Rm) 00040, Italy
| | - Roberto Grassia
- Gastroenterology and Digestive Endoscopy Unit, ASST Cremona, Cremona 26100, Italy
| |
Collapse
|
19
|
Impact of tumor size and location on endoscopic ultrasound-guided sampling of pancreatic neuroendocrine tumors: A recursive partitioning analysis. Pancreatology 2022; 22:644-650. [PMID: 35589512 DOI: 10.1016/j.pan.2022.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/20/2022] [Accepted: 04/26/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND Current guidelines provide weak recommendations to treat small (<2 cm) non-functional pancreatic neuroendocrine tumors with low Ki-67 proliferation index either by resection or clinical follow-up. However, there is a lack of consensus regarding the minimal size of pNET, which allows EUS-guided biopsy with high enough diagnostic accuracy for stratification. METHODS We conducted a retrospective, bicentric analysis of patients who had undergone EUS-guided pNET sampling in two tertiary care Endoscopy Units in Germany and Poland. Using a recursive partitioning of the tree-aided model, we aimed to stratify the probability of successful EUS-guided biopsy of pNET lesions according to their size and location. RESULTS In our pNET cohort, successful histological confirmation of a pNET diagnosis was achieved in 59/69 (85.5%) cases at the initial EUS-guided biopsy. In 41 patients with a pNET size less than 18.5 mm, the EUS-guided first biopsy was successful in 90.2%. In 16 of these patients with smaller lesions, EUS-guided sampling was 100% in very small (less than 11 mm) and extremely small lesions (less than 8 mm). The biopsy success rate was 100% in tail lesions in the size range between ≥5.95 and <8.1 mm but only 33.3% independent of the investigator in pancreatic head or body, with an error rate of 11.2% CONCLUSION: Using a recursive partitioning of the tree-aided stratification model, we demonstrate for the first time that in balancing risks and benefits, very small pNETs (<1 cm) in the tail of the pancreas should be sampled under EUS-guidance.
Collapse
|
20
|
Alghamdi A, Palmieri V, Alotaibi N, Barkun A, Zogopoulos G, Chaudhury P, Barkun J, Miller C, Benmassaoud A, Parent J, Martel M, Chen YI. Preoperative Endoscopic Ultrasound Fine Needle Aspiration Versus Upfront Surgery in Resectable Pancreatic Cancer: A Systematic Review and Meta-analysis of Clinical Outcomes Including Survival and Risk of Tumor Recurrence. J Can Assoc Gastroenterol 2022; 5:121-128. [PMID: 35669844 PMCID: PMC9157295 DOI: 10.1093/jcag/gwab037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 09/04/2021] [Indexed: 11/15/2022] Open
Abstract
Background and Aim Endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) is the standard of care in advanced pancreatic cancer. Its role in resectable disease, however, is controversial. This meta-analysis aims to ascertain the clinical outcomes of patients with resectable pancreatic cancer undergoing preoperative EUS-FNA compared to those going directly to surgery. Methods A literature search was performed from 1996 to April 2019 using MEDLINE, EMBASE, and ISI Web of Knowledge for studies comparing preoperative EUS-FNA to EUS without FNA in resectable pancreatic cancer for clinical outcomes. The primary outcome is overall survival (OS). Secondary outcomes include cancer-free survival, tumor recurrence and peritoneal carcinomatosis, and post-FNA-pancreatitis rate. Results Six retrospective studies were included. Preoperative EUS-FNA had better OS than the non-FNA group (WMD, 4.40 months [0.02 to 8.78]). Cancer-free survival did not differ significantly between the two groups (WMD, 2.08 months [-2.22 to 6.38]). EUS with FNA was not associated with increased rates of tumor recurrence or peritoneal carcinomatosis. Conclusion Preoperative EUS-FNA in resectable pancreatic cancer may be associated with significantly greater OS when compared to the non-FNA group, with no significant difference in the rates of tumor recurrence or peritoneal seeding. Important limitations of our meta-analysis include the lack of prospective controlled data, which are unlikely to emerge given feasible constraints.
Collapse
Affiliation(s)
- Adel Alghamdi
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Vincent Palmieri
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Nawaf Alotaibi
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Alan Barkun
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - George Zogopoulos
- Department of Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Prosanto Chaudhury
- Department of Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Jeffrey Barkun
- Department of Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Corey Miller
- Division of Gastroenterology and Hepatology, Jewish General Hospital, Montreal, Quebec, Canada
| | - Amine Benmassaoud
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Josee Parent
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Myriam Martel
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Yen-I Chen
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
21
|
Dao BT, Nguyen TV, Pham HH, Nguyen HQ. Phase recognition in contrast‐enhanced CT scans based on deep learning and random sampling. Med Phys 2022; 49:4518-4528. [DOI: 10.1002/mp.15551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 01/17/2022] [Accepted: 01/31/2022] [Indexed: 12/19/2022] Open
Affiliation(s)
- Binh T. Dao
- Smart Health Center VinBigData JSC Hanoi Vietnam
| | | | - Hieu H. Pham
- Smart Health Center VinBigData JSC Hanoi Vietnam
- College of Engineering & Computer Science VinUniversity Hanoi Vietnam
- VinUni‐Illinois Smart Health Center Hanoi Vietnam
| | - Ha Q. Nguyen
- Smart Health Center VinBigData JSC Hanoi Vietnam
- College of Engineering & Computer Science VinUniversity Hanoi Vietnam
| |
Collapse
|
22
|
Tong T, Gu J, Xu D, Song L, Zhao Q, Cheng F, Yuan Z, Tian S, Yang X, Tian J, Wang K, Jiang T. Deep learning radiomics based on contrast-enhanced ultrasound images for assisted diagnosis of pancreatic ductal adenocarcinoma and chronic pancreatitis. BMC Med 2022; 20:74. [PMID: 35232446 PMCID: PMC8889703 DOI: 10.1186/s12916-022-02258-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/13/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Accurate and non-invasive diagnosis of pancreatic ductal adenocarcinoma (PDAC) and chronic pancreatitis (CP) can avoid unnecessary puncture and surgery. This study aimed to develop a deep learning radiomics (DLR) model based on contrast-enhanced ultrasound (CEUS) images to assist radiologists in identifying PDAC and CP. METHODS Patients with PDAC or CP were retrospectively enrolled from three hospitals. Detailed clinicopathological data were collected for each patient. Diagnoses were confirmed pathologically using biopsy or surgery in all patients. We developed an end-to-end DLR model for diagnosing PDAC and CP using CEUS images. To verify the clinical application value of the DLR model, two rounds of reader studies were performed. RESULTS A total of 558 patients with pancreatic lesions were enrolled and were split into the training cohort (n=351), internal validation cohort (n=109), and external validation cohorts 1 (n=50) and 2 (n=48). The DLR model achieved an area under curve (AUC) of 0.986 (95% CI 0.975-0.994), 0.978 (95% CI 0.950-0.996), 0.967 (95% CI 0.917-1.000), and 0.953 (95% CI 0.877-1.000) in the training, internal validation, and external validation cohorts 1 and 2, respectively. The sensitivity and specificity of the DLR model were higher than or comparable to the diagnoses of the five radiologists in the three validation cohorts. With the aid of the DLR model, the diagnostic sensitivity of all radiologists was further improved at the expense of a small or no decrease in specificity in the three validation cohorts. CONCLUSIONS The findings of this study suggest that our DLR model can be used as an effective tool to assist radiologists in the diagnosis of PDAC and CP.
Collapse
Affiliation(s)
- Tong Tong
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jionghui Gu
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- Department of Ultrasound, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Dong Xu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), No.1 East Banshan Road, Gongshu District, Hangzhou, 310022, China
| | - Ling Song
- Department of ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiyu Zhao
- Department of Ultrasound, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Fang Cheng
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), No.1 East Banshan Road, Gongshu District, Hangzhou, 310022, China
| | - Zhiqiang Yuan
- Department of ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuyuan Tian
- Department of Ultrasound, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Xin Yang
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Beihang University, Beijing, 100191, China.
| | - Kun Wang
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Tian'an Jiang
- Department of Ultrasound, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China.
- Zhejiang Provincial Key Laboratory of Pulsed Electric Field Technology for Medical Transformation, Hangzhou, 310003, China.
| |
Collapse
|
23
|
Cao Y, Zhao R, Guo K, Ren S, Zhang Y, Lu Z, Tian L, Li T, Chen X, Wang Z. Potential Metabolite Biomarkers for Early Detection of Stage-I Pancreatic Ductal Adenocarcinoma. Front Oncol 2022; 11:744667. [PMID: 35127469 PMCID: PMC8807510 DOI: 10.3389/fonc.2021.744667] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 12/31/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND & OBJECTIVES Pancreatic ductal adenocarcinoma remains an extremely malignant tumor having a poor prognosis. The 5-year survival rate of PDAC is related to its stage (about 80% for stage I vs 20% for other stages). However, detection of PDAC in an early stage is difficult due to the lack of effective screening methods. In this study, we aimed to construct a novel metabolic model for stage-I PDAC detection, using both serum and tissue samples. METHODS We employed an untargeted technique, UHPLC-Q-TOF-MS, to identify the potential metabolite, and then used a targeted technique, GC-TOF-MS, to quantitatively validate. Multivariate and univariate statistics were performed to analyze the metabolomic profiles between stage-I PDAC and healthy controls, including 90 serum and 53 tissue samples. 28 patients with stage-I PDAC and 62 healthy controls were included in this study. RESULTS A total of 10 potential metabolites presented the same expression levels both in serum and in tissue. Among them, a 2-metabolites-model (isoleucine and adrenic acid) for stage-I PDAC was constructed. The area under the curve (AUC) value was 0.93 in the discovery set and 0.90 in the independent validation set. Especially, the serum metabolite model had a better diagnostic performance than CA19-9 (AUC = 0.79). Pathway analysis revealed 11 altered pathways in both serum and tissue of stage-I PDAC. CONCLUSIONS This study developed a novel serum metabolites model that could early separate stage-I PDAC from healthy controls.
Collapse
Affiliation(s)
- Yingying Cao
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Rui Zhao
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Kai Guo
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuai Ren
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yaping Zhang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zipeng Lu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Tian
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Li
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao Chen
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhongqiu Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
24
|
Low kV Computed Tomography of Parenchymal Abdominal Organs-A Systematic Animal Study of Different Contrast Media Injection Protocols. Tomography 2021; 7:815-828. [PMID: 34941641 PMCID: PMC8705800 DOI: 10.3390/tomography7040069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/17/2021] [Accepted: 11/23/2021] [Indexed: 01/14/2023] Open
Abstract
Objectives: To evaluate multiphase low kV computed tomography (CT) imaging of the abdomen with reduced contrast media (CM) dose using different injection protocols. Methods: Two injection protocols were evaluated for use with low kV (80 kV) multiphase abdominal imaging in comparison to the standard procedure acquired at 120 kV (500 mgI/kg; 5 mL/s). This evaluation was conducted in a highly standardized animal study (5 Goettingen minipigs). The low kV protocols consisted of (a) a single-flow (SF) injection with 40% reduced CM dose and injection rate (300 mgI/kg; 3 mL/s) and (b) a DualFlow (DF) injection protocol consisting of 60%/40% contrast to saline ratio administered at 5 mL/s. Dynamic CT was first performed within representative liver regions to determine optimal contrast phases, followed by evaluation of the three protocols in multiphase abdominal CT imaging. The evaluation criteria included contrast enhancement (CE) of abdominal organs and vasculature. Results: The 80 kV DF injection protocol showed similar CE of the abdominal parenchymatous organs and vessels to the 120 kV reference and the 80 kV SF protocol. Hepatic parenchyma showed comparable CT values for all contrast phases. In particular, in the portal venous parenchymal phase, the 80 kV DF protocol demonstrated higher hepatic parenchymal enhancement; however, results were statistically non-significant. Similarly, CE of the kidney, pancreas, and abdominal arterial/venous vessels showed no significant differences between injection protocols. Conclusions: Adapted SF and DF injection protocols with reduced IDR/iodine load offer the potential to calibrate optimal CM doses to the tube voltage in abdominal multiphase low kV CT imaging. The data suggest that the DF approach allows the use of predefined injection protocols and adaption of the contrast to saline ratio to an individualized kV setting and yields the potential for patient-individualized CM adaption.
Collapse
|
25
|
Vernuccio F, Messina C, Merz V, Cannella R, Midiri M. Resectable and Borderline Resectable Pancreatic Ductal Adenocarcinoma: Role of the Radiologist and Oncologist in the Era of Precision Medicine. Diagnostics (Basel) 2021; 11:2166. [PMID: 34829513 PMCID: PMC8623921 DOI: 10.3390/diagnostics11112166] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/22/2021] [Accepted: 11/19/2021] [Indexed: 12/24/2022] Open
Abstract
The incidence and mortality of pancreatic ductal adenocarcinoma are growing over time. The management of patients with pancreatic ductal adenocarcinoma involves a multidisciplinary team, ideally involving experts from surgery, diagnostic imaging, interventional endoscopy, medical oncology, radiation oncology, pathology, geriatric medicine, and palliative care. An adequate staging of pancreatic ductal adenocarcinoma and re-assessment of the tumor after neoadjuvant therapy allows the multidisciplinary team to choose the most appropriate treatment for the patient. This review article discusses advancement in the molecular basis of pancreatic ductal adenocarcinoma, diagnostic tools available for staging and tumor response assessment, and management of resectable or borderline resectable pancreatic cancer.
Collapse
Affiliation(s)
- Federica Vernuccio
- Radiology Unit, University Hospital "Paolo Giaccone", 90127 Palermo, Italy
| | - Carlo Messina
- Oncology Unit, A.R.N.A.S. Civico, 90127 Palermo, Italy
| | - Valeria Merz
- Department of Medical Oncology, Santa Chiara Hospital, 38122 Trento, Italy
| | - Roberto Cannella
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University Hospital of Palermo, Via del Vespro 129, 90127 Palermo, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Via del Vespro 129, 90127 Palermo, Italy
| | - Massimo Midiri
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University Hospital of Palermo, Via del Vespro 129, 90127 Palermo, Italy
| |
Collapse
|
26
|
Yang J, Xu R, Wang C, Qiu J, Ren B, You L. Early screening and diagnosis strategies of pancreatic cancer: a comprehensive review. Cancer Commun (Lond) 2021; 41:1257-1274. [PMID: 34331845 PMCID: PMC8696234 DOI: 10.1002/cac2.12204] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/15/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer is a highly malignant digestive system tumor with a poor prognosis. Most pancreatic cancer patients are diagnosed at an advanced stage or even metastasis due to its highly aggressive characteristics and lack of typical early symptoms. Thus, an early diagnosis of pancreatic cancer is crucial for improving its prognosis. Currently, screening is often applied in high‐risk individuals to achieve the early diagnosis of pancreatic cancer. Fully understanding the risk factors of pancreatic cancer and pathogenesis could help us identify the high‐risk population and achieve early diagnosis and timely treatment of pancreatic cancer. Notably, accumulating studies have been undertaken to improve the detection rate of different imaging methods and the diagnostic accuracy of endoscopic ultrasound‐guided fine‐needle aspiration (EUS‐FNA) which is the golden standard for pancreatic cancer diagnosis. In addition, there are currently no biomarkers with sufficient sensitivity and specificity for the diagnosis of pancreatic cancer to be applied in the clinic. As the only serum biomarker approved by the United States Food and Drug Administration, carbohydrate antigen 19‐9 (CA19‐9) is not recommended to be used in the early screening of pancreatic cancer because of its limited specificity. Recently, increasing numbers of studies focused on the discovering of novel serum biomarkers and exploring their combination with CA19‐9 in the detection of pancreatic cancer. Besides, the application of liquid biopsy involving circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), microRNAs (miRNAs), and exosomes in blood and biomarkers in urine, and saliva in pancreatic cancer diagnosis are drawing more and more attention. Furthermore, many innovative technologies such as artificial intelligence, computer‐aided diagnosis system, metabolomics technology, ion mobility spectrometry (IMS) associated technologies, and novel nanomaterials have been tested for the early diagnosis of pancreatic cancer and have shown promising prospects. Hence, this review aims to summarize the recent progress in the development of early screening and diagnostic methods, including imaging, pathological examination, serological examination, liquid biopsy, as well as other potential diagnostic strategies for pancreatic cancer.
Collapse
Affiliation(s)
- Jinshou Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, P. R. China
| | - Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, P. R. China
| | - Chengcheng Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, P. R. China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, P. R. China
| | - Bo Ren
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, P. R. China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
27
|
Screening for pancreatic cancer: a review for general clinicians. ACTA ACUST UNITED AC 2021; 58:119-128. [PMID: 32364522 DOI: 10.2478/rjim-2020-0009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer (PC) is an exceptionally lethal malignancy with increasing incidence and mortality worldwide. One of the principal challenges in the treatment of PC is that the diagnosis is usually made at a late stage when potentially curative surgical resection is no longer an option. General clinicians including internists and family physicians are well positioned to identify high-risk individuals and refer them to centers with expertise in PC screening and treatment where screening modalities can be employed. Here, we provide an up-to-date review of PC precursor lesions, epidemiology, and risk factors to empower the general clinician to recognize high-risk patients and employ risk reduction strategies. We also review current screening guidelines and modalities and preview progress that is being made to improve screening tests and biomarkers. It is our hope that this review article will empower the general clinician to understand which patients need to be screened for PC, strategies that may be used to reduce PC risk, and which screening modalities are available in order to diminish the lethality of PC.
Collapse
|
28
|
Zeeshan MS, Ramzan Z. Current controversies and advances in the management of pancreatic adenocarcinoma. World J Gastrointest Oncol 2021; 13:472-494. [PMID: 34163568 PMCID: PMC8204360 DOI: 10.4251/wjgo.v13.i6.472] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/22/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic adenocarcinoma is a lethal disease with a mortality rate that has not significantly improved over decades. This is likely due to several challenges unique to pancreatic cancer. Most patients with pancreatic cancer are diagnosed at a late stage of disease due to the lack of specific symptoms prompting an early investigation. A small subset of patients who are diagnosed at an early stage have a better chance at survival with curative surgical resection, but most patients still succumb to the disease in a few years. The dismal overall prognosis is due to suspected micro-metastasis at an early stage. Due to this reason, there is a recent interest in treating all patients with pancreatic cancers with systemic therapy upfront (including the ones that are surgically resectable). This approach is still not the standard of care due to the lack of robust prospective data available. Recent advancements in treatment regimens of chemotherapy, radiation and immunotherapy have improved the overall short-term survival but the long-term survival still remains poor. Novel approaches in diagnosis and treatment have shown promise in clinical studies but long-term clinical data is lacking. The following manuscript presents an overview of the epidemiology, diagnosis, staging, recent advances, novel approaches and controversies in the management of pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Muhammad Shehroz Zeeshan
- Gastrointestinal Section, Department of Medicine, Texas Health Harris Methodist Hospital, Fort Worth, TX 76104, United States
| | - Zeeshan Ramzan
- Gastrointestinal Section, Department of Medicine, Texas Health Harris Methodist Hospital, Fort Worth, TX 76104, United States
| |
Collapse
|
29
|
Ammendola M, Currò G, Laface C, Zuccalà V, Memeo R, Luposella F, Laforgia M, Zizzo N, Zito A, Loisi D, Patruno R, Milella L, Ugenti I, Porcelli M, Navarra G, Gadaleta CD, Ranieri G. Mast Cells Positive for c-Kit Receptor and Tryptase Correlate with Angiogenesis in Cancerous and Adjacent Normal Pancreatic Tissue. Cells 2021; 10:444. [PMID: 33669751 PMCID: PMC7923170 DOI: 10.3390/cells10020444] [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] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/16/2021] [Accepted: 02/16/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Mast cells (MCs) contain proangiogenic factors, in particular tryptase, associated with increased angiogenesis in several tumours. With special reference to pancreatic cancer, few data have been published on the role of MCs in angiogenesis in both pancreatic ductal adenocarcinoma tissue (PDAT) and adjacent normal tissue (ANT). In this study, density of mast cells positive for c-Kit receptor (MCDP-c-KitR), density of mast cells positive for tryptase (MCDPT), area of mast cells positive for tryptase (MCAPT), and angiogenesis in terms of microvascular density (MVD) and endothelial area (EA) were evaluated in a total of 45 PDAT patients with stage T2-3N0-1M0. RESULTS For each analysed tissue parameter, the mean ± standard deviation was evaluated in both PDAT and ANT and differences were evaluated by Student's t-test (p ranged from 0.001 to 0.005). Each analysed tissue parameter was then correlated to each other one by Pearson t-test analysis (p ranged from 0.01 to 0.03). No other correlation among MCDP-c-KitR, MCDPT, MCAPT, MVD, EA and the main clinical-pathological characteristics was found. CONCLUSIONS Our results suggest that tissue parameters increased from ANT to PDAT and that mast cells are strongly associated with angiogenesis in PDAT. On this basis, the inhibition of MCs through tyrosine kinase inhibitors, such as masitinib, or inhibition of tryptase by gabexate mesylate may become potential novel antiangiogenetic approaches in pancreatic cancer therapy.
Collapse
Affiliation(s)
- Michele Ammendola
- Department of Health Science, Digestive Surgery Unit, Medical School, University “Magna Graecia”, Viale Europa, Germaneto, 88100 Catanzaro, Italy; (M.A.); (G.C.); (L.M.); (I.U.)
| | - Giuseppe Currò
- Department of Health Science, Digestive Surgery Unit, Medical School, University “Magna Graecia”, Viale Europa, Germaneto, 88100 Catanzaro, Italy; (M.A.); (G.C.); (L.M.); (I.U.)
| | - Carmelo Laface
- Interventional Oncology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (C.L.); (M.P.); (C.D.G.)
- Department of Biomedical Sciences and Clinical Oncology, Section of Oncology, University of Bari ′Aldo Moro′, 70124 Bari, Italy
| | - Valeria Zuccalà
- Pathology Unit, “Pugliese-Ciaccio” Hospital, Viale Pio X°, 88100 Catanzaro, Italy;
| | - Riccardo Memeo
- Department of Emergency and Organ Transplantation, University Aldo Moro of Bari, 70124 Bari, Italy;
| | - Francesco Luposella
- Direction Départementale de la Cohésion Sociale et de la Protection des Populations des VOSGES (DDCSPP88), 88080 Vittel, France;
| | - Mariarita Laforgia
- Pharmacy Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy;
| | - Nicola Zizzo
- Chair of Pathology, Veterinary Medical School, University “Aldo Moro” of Bari, Via Casamassima, 70010 Bari, Italy; (N.Z.); (R.P.)
| | - Alfredo Zito
- Pathology Unit, National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.Z.); (D.L.)
| | - Donato Loisi
- Pathology Unit, National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.Z.); (D.L.)
| | - Rosa Patruno
- Chair of Pathology, Veterinary Medical School, University “Aldo Moro” of Bari, Via Casamassima, 70010 Bari, Italy; (N.Z.); (R.P.)
| | - Lucia Milella
- Department of Health Science, Digestive Surgery Unit, Medical School, University “Magna Graecia”, Viale Europa, Germaneto, 88100 Catanzaro, Italy; (M.A.); (G.C.); (L.M.); (I.U.)
| | - Ippazio Ugenti
- Department of Health Science, Digestive Surgery Unit, Medical School, University “Magna Graecia”, Viale Europa, Germaneto, 88100 Catanzaro, Italy; (M.A.); (G.C.); (L.M.); (I.U.)
- Department of Emergency and Organ Transplantation, University Aldo Moro of Bari, 70124 Bari, Italy;
| | - Mariangela Porcelli
- Interventional Oncology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (C.L.); (M.P.); (C.D.G.)
| | - Giuseppe Navarra
- Department of Human Pathology of Adult and Evolutive Age, Surgical Oncology Division, University Hospital of Messina, 98100 Messina, Italy;
| | - Cosmo Damiano Gadaleta
- Interventional Oncology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (C.L.); (M.P.); (C.D.G.)
| | - Girolamo Ranieri
- Interventional Oncology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (C.L.); (M.P.); (C.D.G.)
| |
Collapse
|
30
|
Role of Endoscopic Ultrasonography and Endoscopic Retrograde Cholangiopancreatography in the Diagnosis of Pancreatic Cancer. Diagnostics (Basel) 2021; 11:diagnostics11020238. [PMID: 33557084 PMCID: PMC7913831 DOI: 10.3390/diagnostics11020238] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer has the poorest prognosis among all cancers, and early diagnosis is essential for improving the prognosis. Along with radiologic modalities, such as computed tomography (CT) and magnetic resonance imaging (MRI), endoscopic modalities play an important role in the diagnosis of pancreatic cancer. This review evaluates the roles of two of those modalities, endoscopic ultrasonography (EUS) and endoscopic retrograde cholangiopancreatography (ERCP), in the diagnosis of pancreatic cancer. EUS can detect pancreatic cancer with higher sensitivity and has excellent sensitivity for the diagnosis of small pancreatic cancer that cannot be detected by other imaging modalities. EUS may be useful for the surveillance of pancreatic cancer in high-risk individuals. Contrast-enhanced EUS and EUS elastography are also useful for differentiating solid pancreatic tumors. In addition, EUS-guided fine needle aspiration shows excellent sensitivity and specificity, even for small pancreatic cancer, and is an essential examination method for the definitive pathological diagnosis and treatment decision strategy. On the other hand, ERCP is invasive and performed less frequently for the purpose of diagnosing pancreatic cancer. However, ERCP is essential in cases that require evaluation of pancreatic duct stricture that may be early pancreatic cancer or those that require differentiation from focal autoimmune pancreatitis.
Collapse
|
31
|
Mohan SL, Chakkalakkoombil SV, Devaraj SK, Kashyap R, Ganesh RN. The Elusive Appearance of Hyperenhancing Solid Serous Cystadenoma of the Pancreas—Radio-Pathological Correlation. JOURNAL OF GASTROINTESTINAL AND ABDOMINAL RADIOLOGY 2021. [DOI: 10.1055/s-0040-1721293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
AbstractSerous cystadenomas account for ~10 to 29% of pancreatic cystic tumors. Solid serous cystadenoma (SSCA) is a rare variant, first described in 1996, with imaging characteristics different from the classical serous cystadenoma of the pancreas and can cause a diagnostic dilemma due to its resemblance to other solid tumors of the pancreas. To the best of our knowledge, only 22 cases of SSCA of pancreas have been reported till date.A 50-year-old female patient underwent contrast-enhanced computed tomography (CECT) of the abdomen for a hypoechoic lesion detected in the body of the pancreas during ultrasound (US) examination. Due to the hyperenhancement of the well-circumscribed lesion in the arterial phase, a provisional diagnosis of neuroendocrine tumor was considered. Gallium 68-labeled somatostatin-analog (Ga 68-DOTANOC) positron emission tomography CT scan did not show any uptake within the lesion and endoscopic US (EUS)-guided fine-needle aspiration cytology (FNAC) was also inconclusive. She underwent laparotomy and the lesion was enucleated and it was proven to be a serous microcystadenoma on postoperative histopathologic examination.A diagnosis of SSCA should be considered for solid-appearing pancreatic lesions with characteristic CECT features such as arterial phase hyperenhancement, and immediate washout, along with negative results on DOTANOC scan and EUS-guided FNAC. Malignant transformation of SSCA has not been reported till date, and hence these can be safely followed up, instead of invasive surgery.
Collapse
Affiliation(s)
- Supraja Laguduva Mohan
- Department of Radiodiagnosis, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | | | - Sunil Kumar Devaraj
- Department of Radiodiagnosis, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Ravindar Kashyap
- Department of Radiodiagnosis, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Rajesh Nachiappa Ganesh
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| |
Collapse
|
32
|
Jugniot N, Bam R, Meuillet EJ, Unger EC, Paulmurugan R. Current status of targeted microbubbles in diagnostic molecular imaging of pancreatic cancer. Bioeng Transl Med 2021; 6:e10183. [PMID: 33532585 PMCID: PMC7823123 DOI: 10.1002/btm2.10183] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/19/2020] [Accepted: 08/19/2020] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is often associated with a poor prognosis due to silent onset, resistance to therapies, and rapid spreading. Most patients are ineligible for curable surgery as they present with advanced disease at the time of diagnosis. Present diagnostic methods relying on anatomical changes have various limitations including difficulty to discriminate between benign and malignant conditions, invasiveness, the ambiguity of imaging results, or the inability to detect molecular biomarkers of PDAC initiation and progression. Therefore, new imaging technologies with high sensitivity and specificity are critically needed for accurately detecting PDAC and noninvasively characterizing molecular features driving its pathogenesis. Contrast enhanced targeted ultrasound (CETUS) is an upcoming molecular imaging modality that specifically addresses these issues. Unlike anatomical imaging modalities such as CT and MRI, molecular imaging using CETUS is promising for early and accurate detection of PDAC. The use of molecularly targeted microbubbles that bind to neovascular targets can enhance the ultrasound signal specifically from malignant PDAC tissues. This review discusses the current state of diagnostic imaging modalities for pancreatic cancer and places a special focus on ultrasound targeted-microbubble technology together with its clinical translatability for PDAC detection.
Collapse
Affiliation(s)
- Natacha Jugniot
- Department of RadiologyMolecular Imaging Program at Stanford, Stanford UniversityPalo AltoCaliforniaUSA
| | - Rakesh Bam
- Department of RadiologyMolecular Imaging Program at Stanford, Stanford UniversityPalo AltoCaliforniaUSA
| | | | | | - Ramasamy Paulmurugan
- Department of RadiologyMolecular Imaging Program at Stanford, Stanford UniversityPalo AltoCaliforniaUSA
| |
Collapse
|
33
|
Barakat MT, Banerjee S. Incidental biliary dilation in the era of the opiate epidemic: High prevalence of biliary dilation in opiate users evaluated in the Emergency Department. World J Hepatol 2020; 12:1289-1298. [PMID: 33442455 PMCID: PMC7772725 DOI: 10.4254/wjh.v12.i12.1289] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/12/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Biliary dilation is frequently related to obstruction; however, non-obstructive factors such as age and previous cholecystectomy have also been reported. In the past two decades there has been a dramatic increase in opiate use/dependence and utilization of cross-sectional abdominal imaging, with increased detection of biliary dilation, particularly in patients who use opiates.
AIM To evaluate associations between opiate use, age, cholecystectomy status, ethnicity, gender, and body mass index utilizing our institution’s integrated informatics platform.
METHODS One thousand six hundred and eighty-five patients (20% sample) presenting to our Emergency Department for all causes over a 5-year period (2011-2016) who had undergone cross-sectional abdominal imaging and had normal total bilirubin were included and analyzed.
RESULTS Common bile duct (CBD) diameter was significantly higher in opiate users compared to non-opiate users (8.67 mm vs 7.24 mm, P < 0.001) and in patients with a history of cholecystectomy compared to those with an intact gallbladder (8.98 vs 6.72, P < 0.001). For patients with an intact gallbladder who did not use opiates (n = 432), increasing age did not predict CBD diameter (r2 = 0.159, P = 0.873). Height weakly predicted CBD diameter (r2 = 0.561, P = 0.018), but weight, body mass index, ethnicity and gender did not.
CONCLUSION Opiate use and a history of cholecystectomy are associated with CBD dilation in the absence of an obstructive process. Age alone is not associated with increased CBD diameter. These findings suggest that factors such as opiate use and history of cholecystectomy may underlie the previously-reported association of advancing age with increased CBD diameter. Further prospective study is warranted.
Collapse
Affiliation(s)
- Monique T Barakat
- Divisions of Adult and Pediatric Gastroenterology and Hepatology, Stanford University Medical Center, Stanford, CA 94305, United States
| | - Subhas Banerjee
- Division of Gastroenterology and Hepatology, Stanford University Medical Center, Stanford, CA 94304, United States
| |
Collapse
|
34
|
Tanaka S, Fukuda J, Nakao M, Ioka T, Ashida R, Takakura R, Okagaki S, Katayama K, Ohkawa K, Ikezawa K, Nagata S. Effectiveness of Contrast-Enhanced Ultrasonography for the Characterization of Small and Early Stage Pancreatic Adenocarcinoma. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:2245-2253. [PMID: 32527594 DOI: 10.1016/j.ultrasmedbio.2020.04.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 06/11/2023]
Abstract
The purpose of this retrospective study is to evaluate the effectiveness of contrast-enhanced ultrasonography for the characterization of small and early stage pancreatic adenocarcinoma. Contrast-enhanced ultrasonography and contrast-enhanced computed tomography (CT) were performed in 200 cases, with pancreatic hypoechoic regions detected with ultrasonography. Assuming that hypo-enhancement was indicative of pancreatic adenocarcinoma, the sensitivity of each imaging modality was calculated. The sensitivities of contrast-enhanced ultrasonography and contrast-enhanced CT to characterize adenocarcinoma were 97.0% and 77.0% (p < 0.0001) for all 100 adenocarcinoma cases, 100% and 76.7% (p = 0.0016) for 43 small (≤20 mm) cancers, 100% and 58.3% (p = 0.0253) for 12 smaller (≤10 mm) cancers and 100% and 72.2% (p = 0.0016) for 36 stage IA cancers, respectively. The sensitivity of contrast-enhanced ultrasonography was sufficiently high and significantly superior to that of contrast-enhanced CT. Contrast-enhanced ultrasonography is a sensitive tool for selecting highly possible pancreatic adenocarcinoma lesions without overlooking early stage tiny adenocarcinomas among a large number of hypoechoic lesions detected with ultrasonography.
Collapse
Affiliation(s)
- Sachiko Tanaka
- Department of Gastrointestinal Cancer Screening and Surveillance, Osaka International Cancer Institute, Osaka, Japan.
| | - Junko Fukuda
- Department of Gastrointestinal Cancer Screening and Surveillance, Osaka International Cancer Institute, Osaka, Japan
| | - Miho Nakao
- Department of Gastrointestinal Cancer Screening and Surveillance, Osaka International Cancer Institute, Osaka, Japan
| | - Tatsuya Ioka
- Department of Gastrointestinal Cancer Screening and Surveillance, Osaka International Cancer Institute, Osaka, Japan
| | - Reiko Ashida
- Department of Gastrointestinal Cancer Screening and Surveillance, Osaka International Cancer Institute, Osaka, Japan
| | - Rena Takakura
- Health Administration Center, Sumitomo Hospital, Osaka, Japan
| | - Suetsumi Okagaki
- Department of Gastrointestinal Cancer Screening and Surveillance, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuhiro Katayama
- Department of Gastrointestinal Cancer Screening and Surveillance, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuyoshi Ohkawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kenji Ikezawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Shigenori Nagata
- Department of Diagnostic Pathology and Cytology, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
35
|
Gupta S, Puri SK. Comparative analysis and assessment of diagnostic accuracy of 256 slice CT and endoscopic ultrasound in evaluation of pancreatic masses. Indian J Radiol Imaging 2020; 30:294-303. [PMID: 33273763 PMCID: PMC7694713 DOI: 10.4103/ijri.ijri_437_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/09/2020] [Accepted: 06/03/2020] [Indexed: 11/17/2022] Open
Abstract
CONTEXT Pancreatic masses are routinely encountered on imaging and often present as a diagnostic dilemma. These masses range from benign inflammatory masses, requiring no intervention to malignant masses, which carry grave prognosis and hence require aggressive management. AIMS Compare the diagnostic accuracy of 256 multislice CT and endoscopic ultrasound (EUS) in characterization and assessment of resectability of pancreatic masses and compare the multidetector computed tomography (MDCT) and EUS findings with histopathological findings. SETTINGS AND DESIGN Prospective study. SUBJECTS AND METHODS 36 patients with pancreatic masses were included who underwent dual phase CT using pancreatic protocol and EUS using 5-13 MHz transducer. Fine needle aspiration cytology (FNAC) was done wherever feasible. Parameters regarding tumor size, location, imaging morphology, and vessel involvement were recorded. Findings were compared with histopathological/operative diagnosis/clinical follow-up. STATISTICAL ANALYSIS USED Descriptive statistics with percentages and proportions and Chi-square test. RESULTS Multidetector computed tomography (MDCT) and EUS established diagnosis consistent with tissue diagnosis in 30 (83%) and 22 (61%) patients, respectively. However, the best results were obtained with the combined use of MDCT and EUS. The number of patients categorized as inconclusive by MDCT were lower compared to EUS. Assessing resectability for pancreatic adenocarcinoma, MDCT showed specificity and positive predictive value (PPV) of 100% compared to EUS, which had specificity and PPV of 75% and 92.3%, respectively. MDCT is the first-line imaging modality in detection, characterization of pancreatic masses, and assessment of resectability in malignant neoplasms. EUS is beneficial in the detection of masses <2 cm in size causing pancreatic contour deformity on CT, for guiding FNAC. MDCT and EUS with EUS-guided FNA are complementary not competitive tools in preoperative imaging of pancreatic masses.
Collapse
Affiliation(s)
- Surabhi Gupta
- Department of Radiology, G.B. Pant Institute of Postgraduate Medical Education and Research, Jawahar Lal Nehru Marg, New Delhi, India
| | - Sunil K Puri
- Department of Radiology, G.B. Pant Institute of Postgraduate Medical Education and Research, Jawahar Lal Nehru Marg, New Delhi, India
| |
Collapse
|
36
|
An Immunosensor for the Detection of ULBP2 Biomarker. MICROMACHINES 2020; 11:mi11060568. [PMID: 32503144 PMCID: PMC7344431 DOI: 10.3390/mi11060568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 01/20/2023]
Abstract
Pancreatic cancer (PC) is a global health problem that features a very high mortality rate. The UL16 binding protein 2 (ULBP2) is a new biomarker for PC detection. This study develops a simple, reliable, and inexpensive immunosensor for the detection of the ULBP2 antigen while also investigating the effects of an array configuration of connected sensors and zinc oxide (ZnO) nanoparticles on the immunosensor’s sensitivity. The ULBP2 antibody was immobilized onto the screen-printed carbon electrode (SPCE) surfaces of three different sensors: a simple SPCE (ULBP2-SPCE); an SPCE array, which is a series of identical SPCE connected to each other at different arrangements of rows and columns (ULBP2-SPCE-1x2 and ULBP2-SPCE-1x3); and an SPCE combined with ZnO nanoparticles (ULBP2-ZnO/SPCE). Impedance spectrum measurements for the immunosensors to ULBP2 antigen were conducted and compared. According to the result, the array configurations (ULBP2-SPCE-1x2 and ULBP2-SPCE-1x3) show an improvement of sensitivity compared to the ULBP2-SPCE alone, but the improvement is not as significant as that of the ULBP2-ZnO/SPCE configuration (ULBP2-ZnO/SPCE > ULBP2-SPCE: 18 times larger). The ULBP2-ZnO/SPCE immunosensor has a low limit of detection (1 pg/mL) and a high sensitivity (332.2 Ω/Log(pg/mL)), excellent linearity (R2 = 0.98), good repeatability (coefficients of variation = 5.03%), and is stable in long-term storage (retaining 95% activity after 28 days storage). In an array configuration, the immunosensor has an increased signal-to-noise ratio (ULBP2-SPCE-1x3 > ULBP2-SPCE: 1.5-fold) and sensitivity (ULBP2-SPCE-1x3 > ULBP2-SPCE: 2.6-fold). In conclusion, either the modification with ZnO nanoparticles onto the sensor or the use of an array configuration of sensors can enhance the immunosensor’s sensitivity. In this study, the best immunosensor for detecting ULBP2 antigens is the ULBP2-ZnO/SPCE immunosensor.
Collapse
|
37
|
Elbanna KY, Jang HJ, Kim TK. Imaging diagnosis and staging of pancreatic ductal adenocarcinoma: a comprehensive review. Insights Imaging 2020; 11:58. [PMID: 32335790 PMCID: PMC7183518 DOI: 10.1186/s13244-020-00861-y] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 03/06/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has continued to have a poor prognosis for the last few decades in spite of recent advances in different imaging modalities mainly due to difficulty in early diagnosis and aggressive biological behavior. Early PDAC can be missed on CT due to similar attenuation relative to the normal pancreas, small size, or hidden location in the uncinate process. Tumor resectability and its contingency on the vascular invasion most commonly assessed with multi-phasic thin-slice CT is a continuously changing concept, particularly in the era of frequent neoadjuvant therapy. Coexistent celiac artery stenosis may affect the surgical plan in patients undergoing pancreaticoduodenectomy. In this review, we discuss the challenges related to the imaging of PDAC. These include radiological and clinical subtleties of the tumor, evolving imaging criteria for tumor resectability, preoperative diagnosis of accompanying celiac artery stenosis, and post-neoadjuvant therapy imaging. For each category, the key imaging features and potential pitfalls on cross-sectional imaging will be discussed. Also, we will describe the imaging discriminators of potential mimickers of PDAC.
Collapse
Affiliation(s)
- Khaled Y Elbanna
- Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto, Toronto, ON, Canada.
| | - Hyun-Jung Jang
- Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto, Toronto, ON, Canada
| | - Tae Kyoung Kim
- Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
38
|
Burns EA, Kasparian S, Khan U, Abdelrahim M. Pancreatic adenocarcinoma with early esophageal metastasis: A case report and review of literature. World J Clin Oncol 2020; 11:83-90. [PMID: 32133277 PMCID: PMC7046924 DOI: 10.5306/wjco.v11.i2.83] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 11/23/2019] [Accepted: 12/20/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma is an aggressive malignancy with a high propensity to metastasize. Esophageal metastasis manifesting as dysphagia is rarely reported in the literature and has not to our knowledge been reported prior to the appearance of the primary disease.
CASE SUMMARY A patient presented with progressive dysphagia to solids and a persistent earache. Computed tomography of the neck and chest revealed a 3.0 cm × 1.8 cm heterogeneous mass originating from the upper third of the esophagus, necrotic cervical and supraclavicular lymphadenopathy, and bilateral pulmonary nodules. She underwent a core needle biopsy of a right cervical node, which suggested a well-differentiated adenocarcinoma of unknown primary. She had an upper endoscopy with biopsy of the esophageal mass suggestive of a well-differentiated adenocarcinoma. Positron emission tomography imaging revealed increased uptake in the esophageal mass, cervical, and mediastinal lymph nodes. She was started on folinic acid, fluorouracil, and oxaliplatin. Prior to initiation of cycle 8, the patient was found to have a pancreatic body mass that was not present on prior radiographic imaging, confirmed by endoscopic ultrasonography and biopsy to be pancreatic adenocarcinoma. CA19-9 was > 10000 U/mL, suggesting a primary pancreaticobiliary origin.
CONCLUSION Esophageal metastasis diagnosed before primary pancreatic adenocarcinoma is rare. This case highlights the profound metastatic potential of pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Ethan Alexander Burns
- Department of Internal Medicine, Houston Methodist Hospital, Houston, TX 77030, United States
| | - Saro Kasparian
- Department of Internal Medicine, Houston Methodist Hospital, Houston, TX 77030, United States
| | - Usman Khan
- Department of Oncology, Houston Methodist Cancer Center, Houston, TX 77030, United States
| | - Maen Abdelrahim
- Department of Oncology, Houston Methodist Cancer Center, Houston, TX 77030, United States
| |
Collapse
|
39
|
Haj-Mirzaian A, Kawamoto S, Zaheer A, Hruban RH, Fishman EK, Chu LC. Pitfalls in the MDCT of pancreatic cancer: strategies for minimizing errors. Abdom Radiol (NY) 2020; 45:457-478. [PMID: 31897686 DOI: 10.1007/s00261-019-02390-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Multidetector computed tomography (MDCT) is a widely used cross-sectional imaging modality for initial evaluation of patients with suspected pancreatic ductal adenocarcinoma (PDAC). However, diagnosis of PDAC can be challenging due to numerous pitfalls associated with image acquisition and interpretation, including technical factors, imaging features, and cognitive errors. Accurate diagnosis requires familiarity with these pitfalls, as these can be minimized using systematic strategies. Suboptimal acquisition protocols and other technical errors such as motion artifacts and incomplete anatomical coverage increase the risk of misdiagnosis. Interpretation of images can be challenging due to intrinsic tumor features (including small and isoenhancing masses, exophytic masses, subtle pancreatic duct irregularities, and diffuse tumor infiltration), presence of coexisting pathology (including chronic pancreatitis and intraductal papillary mucinous neoplasm), mimickers of PDAC (including focal fatty infiltration and focal pancreatitis), distracting findings, and satisfaction of search. Awareness of pitfalls associated with the diagnosis of PDAC along with the strategies to avoid them will help radiologists to minimize technical and interpretation errors. Cognizance and mitigation of these errors can lead to earlier PDAC diagnosis and ultimately improve patient prognosis.
Collapse
Affiliation(s)
- Arya Haj-Mirzaian
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Satomi Kawamoto
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Atif Zaheer
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ralph H Hruban
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elliot K Fishman
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Linda C Chu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
40
|
PET in Gastrointestinal, Pancreatic, and Liver Cancers. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
41
|
De Jesus-Acosta A, Narang A, Mauro L, Herman J, Jaffee EM, Laheru DA. Carcinoma of the Pancreas. ABELOFF'S CLINICAL ONCOLOGY 2020:1342-1360.e7. [DOI: 10.1016/b978-0-323-47674-4.00078-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
42
|
Dallongeville A, Corno L, Silvera S, Boulay-Coletta I, Zins M. Initial Diagnosis and Staging of Pancreatic Cancer Including Main Differentials. Semin Ultrasound CT MR 2019; 40:436-468. [PMID: 31806145 DOI: 10.1053/j.sult.2019.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
43
|
El Kayal N, Lennartz S, Ekdawi S, Holz J, Slebocki K, Haneder S, Wybranski C, Mohallel A, Eid M, Grüll H, Persigehl T, Borggrefe J, Maintz D, Heneweer C. Value of spectral detector computed tomography for assessment of pancreatic lesions. Eur J Radiol 2019; 118:215-222. [DOI: 10.1016/j.ejrad.2019.07.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/08/2019] [Accepted: 07/15/2019] [Indexed: 01/05/2023]
|
44
|
Sofuni A, Tsuchiya T, Itoi T. Ultrasound diagnosis of pancreatic solid tumors. J Med Ultrason (2001) 2019; 47:359-376. [PMID: 31420821 DOI: 10.1007/s10396-019-00968-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/16/2019] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Advances and widespread use of various diagnostic imaging modalities have dramatically improved our ability to visualize and diagnose pancreatic diseases. In particular, ultrasonography in pancreatic diseases plays an important role from screening to diagnosis as a simple and safe examination method. METHODS The basic scanning method of transabdominal pancreatic ultrasonography, characterization, and differential diagnosis by ultrasonography including contrast-enhanced ultrasonography (CEUS) for solid pancreatic tumors are reviewed with reference to various papers. RESULTS In recent years, the ability to visualize and diagnose pancreatic mass lesions has been dramatically improved with advances in ultrasound equipment. In particular, CEUS using an ultrasound contrast agent has made it possible to evaluate hemodynamics in organs or lesions as well as in the flow signal of arterial blood vessels, and it has played an important role not only in diagnosis of the presence of a lesion but also in the qualitative diagnosis. The enhancement behavior and pattern with CEUS of pancreatic solid tumors is shown in text and Fig. 9. Moreover, the flow chart for diagnosing pancreatic solid tumors with CEUS classifying the enhancement behavior and pattern for pancreatic solid tumors on CEUS is shown (Fig. 10). In meta-analyses, the pooled sensitivity in the differential diagnosis of pancreatic adenocarcinomas and other pancreatic focal masses with CEUS was 86-90%, and the pooled specificity was 75-88%. CONCLUSION CEUS is a minimally invasive and useful diagnostic method that can be used to make a simple and quick qualitative diagnosis of pancreatic diseases. CEUS provides a lot of information important for diagnosis, and has led to changes in the conventional diagnostic systems in pancreatic diseases.
Collapse
Affiliation(s)
- Atsushi Sofuni
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan.
| | - Takayoshi Tsuchiya
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Takao Itoi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| |
Collapse
|
45
|
Shobassy M, Husainat N, Tabash A, Patel K, El-Serag HB, Othman MO. Endoscopic Ultrasound Findings in Patients Diagnosed with Exocrine Pancreatic Insufficiency by Low Fecal Elastase-1. Gastroenterol Res Pract 2019; 2019:5290642. [PMID: 31485218 PMCID: PMC6710758 DOI: 10.1155/2019/5290642] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/09/2019] [Accepted: 07/02/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND AIMS Fecal elastase-1 (FE-1) as a screening test for exocrine pancreatic insufficiency (EPI) is gaining popularity in clinical practice. The role of imaging in patients with FE-1-related suspicion of EPI remains unclear. The aim of this study was to characterize endoscopic ultrasound (EUS) findings for patients with low FE-1. METHODS A retrospective cross-sectional study was performed in 40 patients who had low FE-1 and underwent EUS to evaluate the pancreas. We obtained data on demographic and lifestyle factors, EUS findings, and histopathology results. We compared these variables between patients with FE-1 < 100 mcg/g vs. 100-200 mcg/g. RESULTS Most patients (82.5%) established one or more new diagnoses from EUS. Diagnoses included: definitive chronic pancreatitis (n = 29, 72.5%), fatty pancreas (n = 9, 22.5%), and pancreatic solid mass or cyst (n = 9, 22.5%). Half (n = 4) of the solid or cystic lesions were neoplastic. All patients with a solid pancreatic mass also had concurrent chronic pancreatitis. There were no significant differences in EUS findings or demographic or lifestyle factors between groups with FE-1 < 100 mcg/g vs. 100-200 mcg/g. CONCLUSION Chronic pancreatitis is the most common EUS finding in patients with low FE-1 levels. EUS appears helpful in determining the cause of EPI in most patients with low FE-1 and may detect unsuspected pancreatic neoplasia.
Collapse
Affiliation(s)
- Mazen Shobassy
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Nedaa Husainat
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Abdalaziz Tabash
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Kalpesh Patel
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Hashem B. El-Serag
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Innovations in Quality, Effectiveness, and Safety, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
- Section of Gastroenterology and Hepatology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Mohamed O. Othman
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
46
|
Okagawa Y, Kondo T, Tsuji Y, Takayama T, Oiwa S, Yoshida M, Ihara H, Sumiyoshi T, Hirayama M, Kondo H. Natural History of Pancreatic Ductal Adenocarcinoma Diagnosed During Observation of Other Organ Cancers. AMERICAN JOURNAL OF CASE REPORTS 2019; 20:1080-1084. [PMID: 31335860 PMCID: PMC6668584 DOI: 10.12659/ajcr.917197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 05/26/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a rapidly progressive malignancy that exhibits an extremely poor prognosis, with most cases diagnosed at an advanced stage. To date, few reports have explored the natural history of PDAC, and the period leading up to the detection of PDAC as a tumor with contrast-enhanced computed tomography (CECT) remains unclear. Here, we report 3 PDAC cases diagnosed incidentally by repeating imaging examinations during observation of other organ cancers. CASE REPORT Two patients were undergoing postoperative follow-up for colorectal cancer; owing to the elevation of serum CA19-9 or dilatation of the main pancreatic duct, both cases were finally diagnosed with PDAC. Another patient was administered neoadjuvant chemotherapy for a gastrointestinal stromal tumor; the fluorodeoxyglucose uptake in the pancreas with fluorodeoxyglucose positron emission tomography for the treatment assessment led to the diagnosis of PDAC. All patients underwent frequent CECT for assessment of other diseases, and PDAC became visible with CECT within 3-4 months of the appearance of indirect findings of PDAC. CONCLUSIONS The period leading up to the detection of PDAC as a tumor with CECT was approximately 3-4 months. These cases suggest that additional imaging examinations should be performed when the indirect findings of PDAC are noted. This report adds value to the literature by elucidating the natural course of PDAC.
Collapse
Affiliation(s)
- Yutaka Okagawa
- Department of Gastroenterology, Tonan Hospital, Sapporo, Hokkaido, Japan
| | - Tomohiro Kondo
- Department of Clinical Oncology, Tonan Hospital, Sapporo, Hokkaido, Japan
| | - Yasushi Tsuji
- Department of Clinical Oncology, Tonan Hospital, Sapporo, Hokkaido, Japan
| | - Toshizo Takayama
- Department of Clinical Oncology, Tonan Hospital, Sapporo, Hokkaido, Japan
| | - Shutaro Oiwa
- Department of Gastroenterology, Tonan Hospital, Sapporo, Hokkaido, Japan
| | - Masahiro Yoshida
- Department of Gastroenterology, Tonan Hospital, Sapporo, Hokkaido, Japan
| | - Hideyuki Ihara
- Department of Gastroenterology, Tonan Hospital, Sapporo, Hokkaido, Japan
| | - Tetsuya Sumiyoshi
- Department of Gastroenterology, Tonan Hospital, Sapporo, Hokkaido, Japan
| | - Michiaki Hirayama
- Department of Gastroenterology, Tonan Hospital, Sapporo, Hokkaido, Japan
| | - Hitoshi Kondo
- Department of Gastroenterology, Tonan Hospital, Sapporo, Hokkaido, Japan
| |
Collapse
|
47
|
Korn RL, Rahmanuddin S, Borazanci E. Use of Precision Imaging in the Evaluation of Pancreas Cancer. Cancer Treat Res 2019; 178:209-236. [PMID: 31209847 DOI: 10.1007/978-3-030-16391-4_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pancreas cancer is an aggressive and fatal disease that will become one of the leading causes of cancer mortality by 2030. An all-out effort is underway to better understand the basic biologic mechanisms of this disease ranging from early development to metastatic disease. In order to change the course of this disease, diagnostic radiology imaging may play a vital role in providing a precise, noninvasive method for early diagnosis and assessment of treatment response. Recent progress in combining medical imaging, advanced image analysis and artificial intelligence, termed radiomics, can offer an innovate approach in detecting the earliest changes of tumor development as well as a rapid method for the detection of response. In this chapter, we introduce the principles of radiomics and demonstrate how it can provide additional information into tumor biology, early detection, and response assessments advancing the goals of precision imaging to deliver the right treatment to the right person at the right time.
Collapse
Affiliation(s)
- Ronald L Korn
- Virginia G Piper Cancer Center at HonorHealth, Scottsdale, AZ, USA. .,Translational Genomics Research Institute, An Affiliate of City of Hope, Phoenix, AZ, USA. .,Imaging Endpoints Core Lab, Scottsdale, AZ, USA.
| | | | - Erkut Borazanci
- Virginia G Piper Cancer Center at HonorHealth, Scottsdale, AZ, USA.,Translational Genomics Research Institute, An Affiliate of City of Hope, Phoenix, AZ, USA
| |
Collapse
|
48
|
Cheng SH, Cheng YJ, Jin ZY, Xue HD. Unresectable pancreatic ductal adenocarcinoma: Role of CT quantitative imaging biomarkers for predicting outcomes of patients treated with chemotherapy. Eur J Radiol 2019; 113:188-197. [PMID: 30927946 DOI: 10.1016/j.ejrad.2019.02.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/04/2019] [Accepted: 02/10/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The primary aim of this study was to determine if computed tomographic (CT) texture analysis measurements of the tumor are independently associated with progression-free survival (PFS) and overall survival (OS) in patients with unresectable pancreatic ductal adenocarcinoma (PDAC), including both unresectable locally advanced and metastatic PDAC, who were treated with chemotherapy. METHODS After an institutional review board waiver was obtained, contrast material-enhanced CT studies in 41 patients with unresectable PDAC who underwent contrast-enhanced CT before chemotherapy between 2014 and 2017 were analyzed in terms of tumor texture, with quantification of mean gray-level intensity (Mean), entropy, mean of positive pixels (MPP), kurtosis, standard deviation (SD), and skewness for fine to coarse textures (spatial scaling factor (SSF) 0-6, respectively). The association between pretreatment and posttreatment texture parameters, as well as Δ value (difference between posttreatment and pretreatment texture parameters), and survival time was assessed by using Cox proportional hazards models and Kaplan-Meier analysis. RESULTS Findings from the multivariate Cox model indicated that tumor size, tumor SD (HR, 0.942; 95% CI: 0.898, 0.988) and skewness (HR, 0.407; 95% CI: 0.172, 0.962) measurements with SSF = 3, and tumor SD (HR, 0.958; 95% CI: 0.92, 0.997) measurements with SSF = 4 were significantly and independently associated with PFS, while tumor size and tumor SD (HR, 0.928; 95% CI: 0.882, 0.976) measurements with SSF = 3 were significantly and independently associated with OS. None of the post-therapy texture parameters or Δ value had a significant association with OS or PFS in multivariate Cox regression models. Medium SD (SSF = 3) of more than 38.38 and coarse SD (SSF = 4) of more than 40.67 were associated with longer PFS after chemotherapy (for SSF = 3, median PFS was 10.0 vs 6.0 months [P = 0.024], and for SSF = 4, median PFS was 12.0 vs 6.0 months [P = 0.003]). SD of 38.38 or greater (SSF = 3) as a dichotomized variable was a significant positive prognostic factor for OS (median OS, 20.0 vs 9.0 months [P = 0.04]). Survival models that included a combination of pretreatment SD (SSF = 3) with tumor size, had the potential to perform better than SD alone, while having no statistical significance in this study (area under the ROC curve, 0.756 vs 0.715 [P = 0.066]). CONCLUSIONS Pretreatment CT quantitative imaging biomarkers from texture analysis are associated with PFS and OS in patients with unresectable PDAC who were treated with chemotherapy, and the combination of pretreatment texture parameters and tumor size have the potential to perform better in survival models than imaging biomarker alone.
Collapse
Affiliation(s)
- Si-Hang Cheng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yue-Juan Cheng
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Zheng-Yu Jin
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Hua-Dan Xue
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
49
|
D'Souza LS, Buscaglia JM. The Use of Endoscopic Ultrasound in the Evaluation of Unexplained Biliary Dilation. Gastrointest Endosc Clin N Am 2019; 29:161-171. [PMID: 30846146 DOI: 10.1016/j.giec.2018.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Isolated biliary dilation, as an incidental diagnosis, is increasing owing to an increase in the use of noninvasive abdominal imaging and poses a diagnostic challenge to physicians especially when further noninvasive diagnostic testing fails to reveal an etiology. This article reviews available data describing the natural history of this clinical scenario and the impact of endoscopic ultrasound examination in the evaluation of unexplained dilation of the common bile duct.
Collapse
Affiliation(s)
- Lionel S D'Souza
- Division of Gastroenterology and Hepatology, Stony Brook University Hospital, 101 Nicolls Road HSC Level 17, Room 60, Stony Brook, NY 11794, USA.
| | - Jonathan M Buscaglia
- Division of Gastroenterology and Hepatology, Stony Brook University Hospital, 101 Nicolls Road HSC Level 17, Room 60, Stony Brook, NY 11794, USA
| |
Collapse
|
50
|
Li J, Li Z, Kan H, Sun Z, Xing J, Cheng Y, Bai C. CA19-9 elevation as an indication to start salvage treatment in surveillance after pancreatic cancer resection. Pancreatology 2019; 19:302-306. [PMID: 30737189 DOI: 10.1016/j.pan.2019.01.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/25/2019] [Accepted: 01/31/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND CA19-9 is the most commonly used tumor marker in the diagnosis, prognosis and surveillance of pancreatic cancer. We hypothesized that CA19-9 elevation can be taken as an indication to start salvage treatment in surveillance after resection. METHODS From January 2014 and July 2017, 80 pancreatic cancer patients who underwent R0 surgical resection and received adjuvant chemotherapy were included. RESULTS Twenty-six (32.5%) patients started salvage treatment at the time of CA19-9 elevation without radiological evidence of recurrence. Fifty-four (67.5%) patients treated conventionally before recurrence was confirmed by radiological examinations. Sixty (75%) patients had CA19-9 elevation that preceded radiographic recurrence by about 3 months. In the intervention group, the median DFS (23.6 months vs. 12.1 months, P < 0.001) and OS (28.1 months vs. 20.7 months, P = 0.049) were significantly longer than those in the control group. CONCLUSIONS CA19-9 elevation could preceded recurrence confirmed by radiographic examinations in most patients. Tumor marker-guided salvage treatment can significantly prolong disease-free survival and overall survival in patients under surveillance after pancreatic cancer resection.
Collapse
Affiliation(s)
- Jiarui Li
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100032, China.
| | - Zhe Li
- Department of Gynecologic Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Haoxuan Kan
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100032, China.
| | - Zhao Sun
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100032, China.
| | - Jiazhang Xing
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100032, China.
| | - Yuejuan Cheng
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100032, China.
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100032, China.
| |
Collapse
|