1
|
Ruff SM, Tsai S. Use of Diagnostic Laparoscopy and Peritoneal Washings for Pancreatic Cancer. Surg Clin North Am 2024; 104:975-985. [PMID: 39237172 DOI: 10.1016/j.suc.2024.05.004] [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: 09/07/2024]
Abstract
Pancreatic adenocarcinoma is an aggressive malignancy that often presents with advanced disease. Accurate staging is essential for treatment planning and shared decision-making with patients. Staging laparoscopy is a minimally invasive procedure that can detect radiographically occult metastatic disease. Its routine use with the collection of peritoneal washings in patients with pancreatic cancer remains controversial. We, herein, review the current literature concerning staging laparoscopy and peritoneal washings in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Samantha M Ruff
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center
| | - Susan Tsai
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center.
| |
Collapse
|
2
|
Gonzalez RS, Yin T, Rosenbaum MW, Quintana L. Evaluation of fine-needle core biopsy specimens for pancreatic ductal adenocarcinoma: Diagnostic utility and helpful histological features. Histopathology 2024; 85:275-284. [PMID: 38659189 DOI: 10.1111/his.15199] [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: 01/29/2024] [Revised: 03/04/2024] [Accepted: 04/06/2024] [Indexed: 04/26/2024]
Abstract
AIMS With the advent of new biopsy devices, fine-needle core biopsy specimens can be obtained from pancreas masses. This study aimed to report the histological spectrum of intrapancreatic adenocarcinoma on fine-needle core biopsy and the accuracy of sampling. METHODS AND RESULTS We identified 423 SharkCore™ fine-needle core biopsies taken from patients with a high clinical concern for pancreatic adenocarcinoma. For each, we recorded patient age and sex, percentage of diagnostic tissue in each sample and tumour site, size and histological findings. The cases came from 392 patients (193 men, 199 women; mean age 69 years). Median diagnostic tissue amount in the samples was 30%. Common histological findings included desmoplasia (36%), single atypical cells (44%), haphazard glandular growth pattern (68%), nuclear pleomorphism > 4:1 (39%), incomplete gland lumens (18%) and detached atypical epithelial strips (37%). Additional levels were ordered on 143 cases. Final clinical diagnoses associated with the 423 cases were adenocarcinoma (n = 343), pancreatitis (n = 22), intraductal neoplasm or other benign/low-grade process (n = 16) and unknown (n = 42, patients lost to follow-up). Of the adenocarcinoma cases, the diagnosis was established by the evaluated fine-needle core biopsy sample alone in 178, by fine-needle aspiration biopsy alone in 30, by both concurrently in 89 and by subsequent biopsy or resection in 37 cases. Among 68 cases called suspicious on fine-needle core biopsy, 78% ultimately represented adenocarcinoma. CONCLUSIONS Fine-needle core biopsy allows for histological diagnosis of pancreatic adenocarcinoma, using known histological parameters. Common findings include single atypical cells, desmoplasia, haphazard gland growth and nuclear pleomorphism. Cases interpreted as suspicious often represent malignancy.
Collapse
Affiliation(s)
- Raul S Gonzalez
- Department of Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, GA, USA
| | - Tiffany Yin
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Matthew W Rosenbaum
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Liza Quintana
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
3
|
Liu B, Huang W, Zhang F, Wang J, Guo J, Huang X, Lei G, Wang J, Ye X, Wang R, Gai B, Hu X, Li M, Li C, Xiao Y, Lin Z, Niu L, Zhu G, Gao F, Niu H, Zhang H, Wu Q, Yang J, Zhao H, Zhang K, Chen Z, Chen T, Zhang H, Wang Z, Li Y. Guidelines for permanent iodine-125 seed interstitial brachytherapy for pancreatic cancer (2023 edition): The Chinese expert consensus workshop report. J Cancer Res Ther 2024; 20:1124-1129. [PMID: 39206973 DOI: 10.4103/jcrt.jcrt_2368_23] [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: 10/31/2023] [Accepted: 04/01/2024] [Indexed: 09/04/2024]
Abstract
ABSTRACT The incidence of pancreatic cancer is increasing worldwide. Approximately, 60% of patients with pancreatic cancer have distant metastases at the time of diagnosis, of which only 10% can be removed using standard resection. Further, patients derive limited benefits from chemotherapy or radiotherapy. As such, alternative methods to achieve local control have emerged, including permanent iodine-125 seed interstitial brachytherapy. In 2023, the Chinese College of Interventionalists, affiliated with the Chinese Medical Doctor Association, organized a group of multi-disciplinary experts to compose guidelines for this treatment modality. The aim of this conference was to standardize the procedure for permanent iodine-125 seed interstitial brachytherapy, including indications, contraindications, pre-procedural preparation, procedural operations, complications, efficacy evaluation, and follow-up.
Collapse
Affiliation(s)
- Bin Liu
- Department of Interventional and Minimal Invasive Oncology, The Second Hospital of Shandong University, Jinan, China
- The Institute of Interventional Oncology, Shandong University, Jinan, China
| | - Wei Huang
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fujun Zhang
- Imaging and Interventional Center, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Junjie Wang
- Department of Radiation Oncology, Cancer Centre, Peking University Third Hospital, Beijing, China
| | - Jinhe Guo
- Department of Radiology, Southeast University, Zhongda Hospital, Nanjing, China
| | - Xuequan Huang
- Department of Interventional Medicine, The First Hospital Affiliated to AMU (Southeast Hospital), Chongqing, China
| | - Guangyan Lei
- Department of Thoracic Surgery, Shaanxi Provincial Cancer Hospital, Xi'an, China
| | - Juan Wang
- Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Xin Ye
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Ruoyu Wang
- Department of Oncology, The Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Baodong Gai
- Department of Gastrointestinal and Colorectal Surgery, The Third Hospital of Jilin University, Changjun, China
| | - Xiaokun Hu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Maoquan Li
- Department of Interventional and Vascular Surgery, Affiliated Tenth People's Hospital of Tongji University, Interventional Vascular Institute of Tongji University, Shanghai, China
| | - Chengli Li
- Department of Radiology, Shandong Provincial Hospital, Jinan, Shandong, China
| | - Yueyong Xiao
- Department of Radiology, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhengyu Lin
- Department of Interventional Therapy, The First Affiliated Hospital of Fujian Medical University, Guang Zhou, Guangzhou, China
| | - Lizhi Niu
- Department of Surgical Oncology, Fuda Cancer Hospital, Guang Zhou, Guangzhou, China
| | - Guangyu Zhu
- Department of Radiology, Southeast University, Zhongda Hospital, Nanjing, China
| | - Fei Gao
- Imaging and Interventional Center, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Hongxin Niu
- Department of Interventional Radiology, Shandong Cancer Hospital, Jinan, China
| | - Hongtao Zhang
- Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Qing Wu
- Department of Oncology, Affiliated Longhua Hospital of Shanghai Traditional Chinese Medicine University, Shanghai, China
| | - Jijin Yang
- Department of Interventional Radiology, Shanghai Changhai Hospital, Shanghai, China
| | - Hong Zhao
- Department of Oncology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Kaixian Zhang
- Department of Oncology, Tengzhou Central People's Hospital, Tengzhou, China
| | - Zhijin Chen
- Department of Interventional Radiology, Luwan Branch of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingsong Chen
- Department of Interventional Radiology, Shanghai Seventh People's Hospital, Shanghai, China
| | - Haoren Zhang
- Present Office, True Health Medical Technology CO., LTD., Zhuhai, China
| | - Zhongmin Wang
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuliang Li
- Department of Interventional and Minimal Invasive Oncology, The Second Hospital of Shandong University, Jinan, China
- The Institute of Interventional Oncology, Shandong University, Jinan, China
| |
Collapse
|
4
|
Di Meo G, Pontrelli A, Testini M, Boggi U. Intrapancreatic common hepatic artery in pancreatoduodenectomy: a technical note on how to deal with this exceedingly rare arterial variation. Updates Surg 2024; 76:1529-1533. [PMID: 38418694 DOI: 10.1007/s13304-024-01784-9] [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: 08/17/2023] [Accepted: 11/20/2023] [Indexed: 03/02/2024]
Abstract
Arterial variations in the liver's blood supply play a pivotal role in the success of pancreatoduodenectomy (PD), impacting both its technical execution and oncological outcomes. Among these variations, a common hepatic artery arising from the superior mesenteric artery (SMA) occurs in about 3% of cases. An exceptionally rare variation is the intrapancreatic common hepatic artery (IPCHA). Preserving or reconstructing the IPCHA is vital during PD to prevent liver and biliary necrosis. Particularly for cases of pancreatic cancer with high rates of intrapancreatic perineural spread, preserving IPCHA without compromising radicality presents challenges. We present a detailed report of the technique used for PD in the presence of IPCHA. Surgical technique details include a pylorus-preserving PD with the Cattell-Braasch maneuver, an artery-first approach, and meticulous dissection using "cold" scissors. We emphasize the importance of strategic surgical planning based on high-quality imaging studies, underscoring the need for pancreatic surgeons to be proficient in managing variations in visceral vessels. In conclusion, this case underscores the significance of navigating rare arterial variations in liver supply during PD, highlighting the necessity for meticulous surgical planning and execution.
Collapse
Affiliation(s)
- Giovanna Di Meo
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari "Aldo Moro", Bari, Italy.
| | - Arianna Pontrelli
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari "Aldo Moro", Bari, Italy
| | - Mario Testini
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari "Aldo Moro", Bari, Italy
| | - Ugo Boggi
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| |
Collapse
|
5
|
Cui Y, Zhao Y, Chen X, Jiang Y, Mao H, Ju S, Peng XG. Value of Non-Contrast-Enhanced Vessel Wall MR Imaging in Assessing Vascular Invasion of Retroperitoneal Tumors. J Magn Reson Imaging 2024; 60:752-764. [PMID: 37929323 DOI: 10.1002/jmri.29120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Due to their location and growth patterns, retroperitoneal tumors often involve the surrounding blood vessels. Clinical decisions on a proper treatment depend on the information on this condition. Evaluation of blood vessels using non-contrast-enhanced vessel wall MRI may provide noninvasive assessment of the extent of tumor invasion to assist clinical decision-making. PURPOSE To investigate the performance and potential of non-contrast-enhanced vessel wall MRI in evaluating the degree of vessel wall invasion of retroperitoneal tumors. STUDY TYPE Prospective. POPULATION Thirty-seven participants (mean age: 60.59 ± 11.77 years, 59% male) with retroperitoneal tumors close to vessels based on their diagnostic computer tomography. FIELD STRENGTH/SEQUENCES 3 T; vessel wall MRI sequences: two-dimensional T2-weighted MultiVane XD turbo spin-echo (2D-T2-MVXD-TSE) and three-dimensional T1-weighted motion sensitized driven equilibrium fat suppression turbo spin-echo (3D-T1-MSDE-TSE) sequences; conventional MRI sequences: T2-weighted fat suppression turbo spin-echo (T2-FS-TSE), T2-weighted turbo spin-echo (T2-TSE), modified Dixon T1-weighted fast field echo (T1-mDixon-FFE), and diffusion-weighted echo planar imaging (DWI-EPI) sequences. ASSESSMENT All patients underwent preoperative imaging using both non-contrast conventional and vessel wall MRI sequences. Images obtained from conventional and vessel wall MRI sequences were evaluated independently by three junior radiologists (3 and 2 years of experience in reading MRI) and reviewed by one senior radiologist (25 years of experience in reading MRI) to assess the degree of vessel wall invasion. MRI were validated results from the clinical standard diagnosis based on surgical confirmation or histopathological reports. Interobserver agreement was determined based on the reports from three readers with similar years of experiences. Intraobserver variability was assessed based on categorizing and recategorizing the vessels of 37 patients 1 month apart. STATISTICAL TESTS Intra-class correlation efficient (ICC), Chi-square test, McNemar test, area under the receiver-operating characteristic curve (AUC), Delong test, P < 0.05 was considered significant. RESULTS The accuracy of vessel wall MRI (91.96%, 95% CI: 85.43-95.71; 103 of 112) in detecting the degree of vessel wall invasion was significantly higher than that of conventional MRI (75%, 95% CI: 66.24-82.10; 84 of 112). The interobserver variability or reproducibility in categorization of the degree of vascular wall invasion was good in evaluating images from conventional and vessel wall MRI sequences (ICC = 0.821, 95% CI: 0.765-0.867 and ICC = 0.881, 95% CI: 0.842-0.913, respectively). DATA CONCLUSION Diagnosis of vessel wall invasion of retroperitoneal tumors and assessment of its severity can be improved by using non-contrast-enhanced vessel wall MRI. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Ying Cui
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Yufei Zhao
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Xiaohui Chen
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Yang Jiang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia, USA
| | - Shenghong Ju
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Xin-Gui Peng
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| |
Collapse
|
6
|
Yasrab M, Thakker S, Wright MJ, Ahmed T, He J, Wolfgang CL, Chu LC, Weiss MJ, Kawamoto S, Johnson PT, Fishman EK, Javed AA. Factors associated with radiological misstaging of pancreatic ductal adenocarcinoma: A retrospective observational study. Curr Probl Diagn Radiol 2024; 53:458-463. [PMID: 38522966 DOI: 10.1067/j.cpradiol.2024.03.001] [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: 11/18/2023] [Accepted: 03/06/2024] [Indexed: 03/26/2024]
Abstract
PURPOSE Accurate staging of disease is vital in determining appropriate care for patients with pancreatic ductal adenocarcinoma (PDAC). It has been shown that the quality of scans and the experience of a radiologist can impact computed tomography (CT) based assessment of disease. The aim of the current study was to evaluate the impact of the rereading of outside hospital (OH) CT by an expert radiologist and a repeat pancreatic protocol CT (PPCT) on staging of disease. METHODS Patients evaluated at the our institute's pancreatic multidisciplinary clinic (2006 to 2014) with OH scan and repeat PPCT performed within 30 days were included. In-house radiologists staged disease using OH scans and repeat PPCT, and factors associated with misstaging were determined. RESULTS The study included 100 patients, with a median time between OH scan and PPCT of 19 days (IQR: 13-23 days.) Stage migration was mostly accounted for by upstaging of disease (58.8 % to 83.3 %) in all comparison groups. When OH scans were rereviewed, 21.5 % of the misstaging was due to missed metastases, however, when rereads were compared to the PPCT, occult metastases accounted for the majority of misstaged patients (62.5 %). Potential factors associated with misstaging were primarily related to imaging technique. CONCLUSION A repeat PPCT results in increased detection of metastatic disease that rereviews of OH scans may otherwise miss. Accessible insurance coverage for repeat PPCT imaging even within 30 days of an OH scan could help optimize delivery of care and alleviate burdens associated with misstaging.
Collapse
Affiliation(s)
- Mohammad Yasrab
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sameer Thakker
- Department of Surgery, New York University Langone Hospital, NYU Langone Health, New York City, NY, USA
| | - Michael J Wright
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Taha Ahmed
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jin He
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher L Wolfgang
- Department of Surgery, New York University Langone Hospital, NYU Langone Health, New York City, NY, USA
| | - Linda C Chu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew J Weiss
- Department of Surgery, Northwell Health, Lake Success, NY, USA
| | - Satomi Kawamoto
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pamela T Johnson
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elliot K Fishman
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ammar A Javed
- Department of Surgery, New York University Langone Hospital, NYU Langone Health, New York City, NY, USA.
| |
Collapse
|
7
|
Cai W, Zhu Y, Teng Z, Li D, Cong R, Chen Z, Ma X, Zhao X. Extracellular volume-based scoring system for tracking tumor progression in pancreatic cancer patients receiving intraoperative radiotherapy. Insights Imaging 2024; 15:116. [PMID: 38735009 PMCID: PMC11089023 DOI: 10.1186/s13244-024-01689-6] [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: 11/14/2023] [Accepted: 04/03/2024] [Indexed: 05/13/2024] Open
Abstract
OBJECTIVES To investigate the value of extracellular volume (ECV) derived from portal-venous phase (PVP) in predicting prognosis in locally advanced pancreatic cancer (LAPC) patients receiving intraoperative radiotherapy (IORT) with initial stable disease (SD) and to construct a risk-scoring system based on ECV and clinical-radiological features. MATERIALS AND METHODS One hundred and three patients with LAPC who received IORT demonstrating SD were enrolled and underwent multiphasic contrast-enhanced CT (CECT) before and after IORT. ECV maps were generated from unenhanced and PVP CT images. Clinical and CT imaging features were analyzed. The independent predictors of progression-free survival (PFS) determined by multivariate Cox regression model were used to construct the risk-scoring system. Time-dependent receiver operating characteristic (ROC) curve analysis and the Kaplan-Meier method were used to evaluate the predictive performance of the scoring system. RESULTS Multivariable analysis revealed that ECV, rim-enhancement, peripancreatic fat infiltration, and carbohydrate antigen 19-9 (CA19-9) response were significant predictors of PFS (all p < 0.05). Time-dependent ROC of the risk-scoring system showed a satisfactory predictive performance for disease progression with area under the curve (AUC) all above 0.70. High-risk patients (risk score ≥ 2) progress significantly faster than low-risk patients (risk score < 2) (p < 0.001). CONCLUSION ECV derived from PVP of conventional CECT was an independent predictor for progression in LAPC patients assessed as SD after IORT. The scoring system integrating ECV, radiological features, and CA19-9 response can be used as a practical tool for stratifying prognosis in these patients, assisting clinicians in developing an appropriate treatment approach. CRITICAL RELEVANCE STATEMENT The scoring system integrating ECV fraction, radiological features, and CA19-9 response can track tumor progression in patients with LAPC receiving IORT, aiding clinicians in choosing individual treatment strategies and improving their prognosis. KEY POINTS Predicting the progression of LAPC in patients receiving IORT is important. Our ECV-based scoring system can risk stratifying patients with initial SD. Appropriate prognostication can assist clinicians in developing appropriate treatment approaches.
Collapse
Affiliation(s)
- Wei Cai
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongjian Zhu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ze Teng
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dengfeng Li
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rong Cong
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhaowei Chen
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohong Ma
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xinming Zhao
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
8
|
Barreto SG, Shrikhande SV, Sirohi B. Neoadjuvant Therapy in Borderline Resectable Pancreatic Cancer. Indian J Surg Oncol 2024; 15:249-254. [PMID: 38817993 PMCID: PMC11133292 DOI: 10.1007/s13193-021-01361-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/31/2021] [Indexed: 12/09/2022] Open
Abstract
In this perspective, we present our assessment of all of the known accumulated evidence on the role of neoadjuvant therapy in the management of borderline resectable pancreatic cancer highlighting the gaps in the data, the current regimens used and providing a brief insight into the way forward.
Collapse
Affiliation(s)
- Savio George Barreto
- Division of Surgery and Perioperative Medicine, Flinders Medical Centre, Bedford Park, Adelaide, South Australia Australia
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia Australia
| | - Shailesh V. Shrikhande
- GI and HPB Services, Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra India
| | - Bhawna Sirohi
- Department of Medical Oncology, Apollo Proton Cancer Centre, Chennai, Tamil Nadu India
| |
Collapse
|
9
|
Muaddi H, Kearse L, Warner S. Multimodal Approaches to Patient Selection for Pancreas Cancer Surgery. Curr Oncol 2024; 31:2260-2273. [PMID: 38668070 PMCID: PMC11049254 DOI: 10.3390/curroncol31040167] [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: 03/24/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
With an overall 5-year survival rate of 12%, pancreas ductal adenocarcinoma (PDAC) is an aggressive cancer that claims more than 50,000 patient lives each year in the United States alone. Even those few patients who undergo curative-intent resection with favorable pathology reports are likely to experience recurrence within the first two years after surgery and ultimately die from their cancer. We hypothesize that risk factors for these early recurrences can be identified with thorough preoperative staging, thus enabling proper patient selection for surgical resection and avoiding unnecessary harm. Herein, we review evidence supporting multidisciplinary and multimodality staging, comprehensive neoadjuvant treatment strategies, and optimal patient selection for curative-intent surgical resections. We further review data generated from our standardized approach at the Mayo Clinic and extrapolate to inform potential future investigations.
Collapse
Affiliation(s)
| | | | - Susanne Warner
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN 55902, USA; (H.M.)
| |
Collapse
|
10
|
Zheng J, He B, Deng L, Zhu X, Li R, Chen K, Zheng C, Wang D, Wang Y, Yu C, Chen G. Prognostic value of diffuse reduction of spleen density on postoperative survival of pancreatic ductal adenocarcinoma: A retrospective study. Asia Pac J Clin Oncol 2024; 20:275-284. [PMID: 36748794 DOI: 10.1111/ajco.13936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 07/05/2022] [Accepted: 01/07/2023] [Indexed: 02/08/2023]
Abstract
PURPOSE It is difficult to predict the prognosis of patients with pancreatic ductal adenocarcinoma (PDAC) before radical operation. The purpose of this study was to explore the connection between the diffuse reduction of spleen density on computed tomography (DROSD) and the postoperative prognosis of patients with PDAC. PATIENTS AND METHODS A total of 160 patients with PDAC who underwent radical surgery in the First Affiliated Hospital of Wenzhou Medical University were enrolled. Cox regression analysis was used to cast the overall survival (OS) and evaluate the prognostic factors. Nomogram was used to forecast the possibility of 1-year, 3-year, and 5-year OS. The prediction accuracy and clinical net benefit are performed by concordance index (C-index), calibration curve, time-dependent receiver operating characteristics (tdROC), and decision curve analysis. RESULTS In multivariable Cox analysis, DROSD is independently related to OS. Advanced age, TNM stage, neutrophil/lymphocyte ratio, and severe complications were also independent prognostic factors. The calibration curves of nomogram showed optimal agreement between prediction and observation. The C-index of nomogram is 0.662 (95%CI, 0.606-0.754). The area under tdROC curve for a 3-year OS of nomogram is 0.770. CONCLUSION DROSD is an independent risk factor for an OS of PDAC. We developed a nomogram that combined imaging features, clinicopathological factors, and systemic inflammatory response to provide a personalized risk assessment for patients with PDAC.
Collapse
Affiliation(s)
- Jiuyi Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Bangjie He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Liming Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xuewen Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Rizhao Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Kaiyu Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Chongming Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Daojie Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yi Wang
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Chang Yu
- Department of Interventional Therapy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
11
|
Lu X, Chen H, Zhang T. Solid pseudopapillary neoplasm (SPN) of the pancreas: current understanding on its malignant potential and management. Discov Oncol 2024; 15:77. [PMID: 38498246 PMCID: PMC10948659 DOI: 10.1007/s12672-024-00905-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 02/22/2024] [Indexed: 03/20/2024] Open
Abstract
Solid pseudopapillary neoplasms (SPN) of the pancreas are presently recognized as low-grade malignant tumors that are frequently observed in young females. This tumor has a low incidence and is associated with an excellent prognosis following surgical resection. Typical SPNs primarily affect the pancreas and tend to have moderate or asymptomatic manifestations. Based on retrospective research, it is anticipated that patients with SPN can achieve disease-free survival, even in cases when metastasis is detected during inspection. However, the incidence of malignant SPN has been consistently underestimated, as evidenced by recent research findings. Malignancy of SPN primarily encompasses invasion and infiltration, metastasis, and recurrence after R0 resection. Imaging technologies such as Ultrasound, Computed Tomography, Magnetic Resonance Imaging, and Position Emission Tomography are capable of preliminarily identifying malignant SPN, which is primarily based on its invasive clinical features. Research on risk factors of malignant SPN revealed that larger tumor size, Ki-67 index, and several other parameters had significant correlations with invasive tumor behavior. Pathologic features of malignant SPNs overlay other pancreatic tumors, nevertheless they can provide valuable assistance in the process of diagnosis. Several confirmed specific pathologic biomarkers are related to its cellular origin, characteristic gene mutation, and cell proliferation. Considering the invasiveness of malignant SPN, it is imperative to enhance the comprehensiveness of its therapy. Tumor resection remains a suggested course of action in line with typical SPN, and additional lymph node dissection is seen as reasonable. Compared to benign SPNs, malignant SPNs have worse prognosis, underscoring the necessity of early identification and treatment in comprehensive medical centers to get improved clinical outcomes.
Collapse
Affiliation(s)
- Xiaoyue Lu
- Peking Union Medical College, Beijing, China
| | - Hao Chen
- Department of General Surgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China.
| |
Collapse
|
12
|
Wang L, Wang G, Wang P, Nie F. Pancreatic ductal adenocarcinoma: CEUS characteristics are correlated with pathological findings and help predict early recurrence after resection. JOURNAL OF CLINICAL ULTRASOUND : JCU 2024; 52:230-240. [PMID: 38018362 DOI: 10.1002/jcu.23622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/30/2023]
Abstract
OBJECTIVES To identify characteristics of preoperative contrast-enhanced ultrasound (CEUS) that could predict early recurrence after curative resection of pancreatic ductal adenocarcinoma (PDAC). METHODS From January 2017 to September 2022, a total of 110 patients with PDAC (all confirmed by samples obtained via operation) who underwent CEUS within 1 month before surgery were enrolled. We proposed five CEUS enhancement patterns (Pattern I, homogeneous enhancement; Pattern II, heterogeneous enhancement without cystic components; pattern III, ring enhancement; Pattern IV, starry enhancement; Pattern V, heterogeneous enhancement with cystic components) of PDAC. Clinical-pathologic and CEUS characteristics for predicting early recurrence (recurrence within 1 year after curative resection) were analyzed. Important CEUS characteristics were compared with the pathological findings. RESULTS Tumor size and TNM stage were closely associated with early recurrence. Incomplete-enhancement and enhancement pattern III, IV and V at CEUS imaging were more prone to early recurrence. Incomplete-enhancement lesions had higher histological tumor grades, less frequent remaining acini, and more frequent necrosis within the tumor. PDACs with pattern I and II had lower histological tumor grades, and pattern III, IV and V had higher histological tumor grades. PDACs with pattern I, II and IV had less frequent intratumoral necrosis than PDACs with pattern III and V, and PDACs with pattern IV had lower MVD values. CONCLUSIONS PDACs with incomplete enhancement and enhancement pattern III, IV and V were more prone to early recurrence after attempted curative resection, and these important CEUS characteristics were closely related to the pathological findings of PDAC.
Collapse
Affiliation(s)
- Lan Wang
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Guojuan Wang
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Peihua Wang
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Fang Nie
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
13
|
Turner KM, Wilson GC, Patel SH, Ahmad SA. ASO Practice Guidelines Series: Management of Resectable, Borderline Resectable, and Locally Advanced Pancreas Cancer. Ann Surg Oncol 2024; 31:1884-1897. [PMID: 37980709 DOI: 10.1245/s10434-023-14585-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: 09/08/2023] [Accepted: 10/29/2023] [Indexed: 11/21/2023]
Abstract
Pancreatic adenocarcinoma is an aggressive disease marked by high rates of both local and distant failure. In the minority of patients with potentially resectable disease, multimodal treatment paradigms have allowed for prolonged survival in an increasingly larger pool of well-selected patients. Therefore, it is critical for surgical oncologists to be abreast of current guideline recommendations for both surgical management and multimodal therapy for pancreas cancer. We discuss these guidelines, as well as the underlying data supporting these positions, to offer surgical oncologists a framework for managing patients with pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Kevin M Turner
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Gregory C Wilson
- Division of Surgical Oncology, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sameer H Patel
- Division of Surgical Oncology, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Syed A Ahmad
- Division of Surgical Oncology, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
14
|
Szczepanski JM, Rudolf MA, Shi J. Clinical Evaluation of the Pancreatic Cancer Microenvironment: Opportunities and Challenges. Cancers (Basel) 2024; 16:794. [PMID: 38398185 PMCID: PMC10887250 DOI: 10.3390/cancers16040794] [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: 01/16/2024] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Advances in our understanding of pancreatic ductal adenocarcinoma (PDAC) and its tumor microenvironment (TME) have the potential to transform treatment for the hundreds of thousands of patients who are diagnosed each year. Whereas the clinical assessment of cancer cell genetics has grown increasingly sophisticated and personalized, current protocols to evaluate the TME have lagged, despite evidence that the TME can be heterogeneous within and between patients. Here, we outline current protocols for PDAC diagnosis and management, review novel biomarkers, and highlight potential opportunities and challenges when evaluating the PDAC TME as we prepare to translate emerging TME-directed therapies to the clinic.
Collapse
Affiliation(s)
| | | | - Jiaqi Shi
- Department of Pathology and Clinical Labs, University of Michigan, Ann Arbor, MI 48109, USA; (J.M.S.); (M.A.R.)
| |
Collapse
|
15
|
Zhao B, Xia C, Xia T, Qiu Y, Zhu L, Cao B, Gao Y, Ge R, Cai W, Ding Z, Yu Q, Lu C, Tang T, Wang Y, Song Y, Long X, Ye J, Lu D, Ju S. Development of a radiomics-based model to predict occult liver metastases of pancreatic ductal adenocarcinoma: a multicenter study. Int J Surg 2024; 110:740-749. [PMID: 38085810 PMCID: PMC10871636 DOI: 10.1097/js9.0000000000000908] [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: 07/19/2023] [Accepted: 11/02/2023] [Indexed: 02/17/2024]
Abstract
BACKGROUND Undetectable occult liver metastases block the long-term survival of pancreatic ductal adenocarcinoma (PDAC). This study aimed to develop a radiomics-based model to predict occult liver metastases and assess its prognostic capacity for survival. MATERIALS AND METHODS Patients who underwent surgical resection and were pathologically proven with PDAC were recruited retrospectively from five tertiary hospitals between January 2015 and December 2020. Radiomics features were extracted from tumors, and the radiomics-based model was developed in the training cohort using LASSO-logistic regression. The model's performance was assessed in the internal and external validation cohorts using the area under the receiver operating curve (AUC). Subsequently, the association of the model's risk stratification with progression-free survival (PFS) and overall survival (OS) was then statistically examined using Cox regression analysis and the log-rank test. RESULTS A total of 438 patients [mean (SD) age, 62.0 (10.0) years; 255 (58.2%) male] were divided into the training cohort ( n =235), internal validation cohort ( n =100), and external validation cohort ( n =103). The radiomics-based model yielded an AUC of 0.73 (95% CI: 0.66-0.80), 0.72 (95% CI: 0.62-0.80), and 0.71 (95% CI: 0.61-0.80) in the training, internal validation, and external validation cohorts, respectively, which were higher than the preoperative clinical model. The model's risk stratification was an independent predictor of PFS (all P <0.05) and OS (all P <0.05). Furthermore, patients in the high-risk group stratified by the model consistently had a significantly shorter PFS and OS at each TNM stage (all P <0.05). CONCLUSION The proposed radiomics-based model provided a promising tool to predict occult liver metastases and had a great significance in prognosis.
Collapse
Affiliation(s)
- Ben Zhao
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Cong Xia
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Tianyi Xia
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Yue Qiu
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Liwen Zhu
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Buyue Cao
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Yin Gao
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Rongjun Ge
- School of Instrument Science and Engineering, Southeast University, Nanjing
| | - Wu Cai
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou
| | - Zhimin Ding
- Department of Radiology, Yijishan Hospital of Wannan Medical College, Wuhu
| | - Qian Yu
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Chunqiang Lu
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Tianyu Tang
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Yuancheng Wang
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Yang Song
- MR Scientific Marketing, Siemens Healthineers, Shanghai
| | - Xueying Long
- Department of Radiology, The Xiangya Hospital of Central South University, Changsha
| | - Jing Ye
- Department of Radiology, Northern Jiangsu People’s Hospital, Yangzhou
| | - Dong Lu
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, People’s Republic of China
| | - Shenghong Ju
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| |
Collapse
|
16
|
Yang HK, Park MS, Han K, Eom G, Chung YE, Choi JY, Bang S, Kang CM, Seong J, Kim MJ. Risk Stratification of Pancreatic Ductal Adenocarcinoma Patients Undergoing Curative-Intent Surgery after Neoadjuvant Therapy. Cancer Res Treat 2024; 56:247-258. [PMID: 37605535 PMCID: PMC10789942 DOI: 10.4143/crt.2023.586] [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: 04/17/2023] [Accepted: 08/21/2023] [Indexed: 08/23/2023] Open
Abstract
PURPOSE Clinical prognostic criteria using preoperative factors were not developed for post-neoadjuvant therapy (NAT) surgery of pancreatic ductal adenocarcinoma (PDAC). We aimed to identify preoperative factors associated with overall survival (OS) in PDAC patients who underwent post-NAT curative-intent surgery and develop risk stratification criteria. MATERIALS AND METHODS Consecutive PDAC patients who underwent post-NAT curative-intent surgeries between 2007 and 2020 were retrospectively analyzed. Demographic, laboratory, surgical, and histopathologic variables were collected. Baseline, preoperative, and interval changes of computed tomography (CT) findings proposed by the Society of Abdominal Radiology and the American Pancreatic Association were analyzed. Cox proportional hazard analysis was used to select preoperative variables associated with OS. We developed risk stratification criteria composed of the significant preoperative variables, i.e., post-NAT response criteria. We compared the discrimination performance of post-NAT response criteria with that of post-NAT pathological (yp) American Joint Cancer Committee TNM staging system. RESULTS One hundred forty-five PDAC patients were included. Stable or increased tumor size on CT (hazard ratio [HR], 2.58; 95% confidence interval [CI], 1.58 to 4.21; p < 0.001) and elevated preoperative carbohydrate antigen 19-9 (CA19-9) level (HR, 1.98; 95% CI, 1.11 to 3.55; p=0.021) were independent factors of OS. The OS of the patient groups stratified by post-NAT response criteria which combined changes in tumor size and CA19-9 showed significant difference (p < 0.001). Such stratification was comparable to ypTNM staging in discrimination performance (difference of C-index, 0.068; 95% CI, -0.012 to 0.142). CONCLUSION "Any degree of decrease in tumor size on CT" and CA19-9 normalization or staying normal were independent favorable factors of OS. The combination of the two factors discriminated OS comparably to ypTNM staging.
Collapse
Affiliation(s)
- Hyun Kyung Yang
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Mi-Suk Park
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyunghwa Han
- Department of Radiology, Research Institute of Radiological Sciences and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Geonsik Eom
- Department of Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Eun Chung
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jin-Young Choi
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seungmin Bang
- Department of Internal Medicine, Severance Hospital, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Chang Moo Kang
- Department of Surgery, Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jinsil Seong
- Department of Radiation Oncology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Myeong-Jin Kim
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
17
|
Hu X, Shi S, Wang Y, Yuan J, Chen M, Wei L, Deng W, Feng ST, Peng Z, Luo Y. Dual-energy CT improves differentiation of non-hypervascular pancreatic neuroendocrine neoplasms from CA 19-9-negative pancreatic ductal adenocarcinomas. LA RADIOLOGIA MEDICA 2024; 129:1-13. [PMID: 37861978 DOI: 10.1007/s11547-023-01733-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/28/2023] [Indexed: 10/21/2023]
Abstract
PURPOSE To evaluate the utility of dual-energy CT (DECT) in differentiating non-hypervascular pancreatic neuroendocrine neoplasms (PNENs) from pancreatic ductal adenocarcinomas (PDACs) with negative carbohydrate antigen 19-9 (CA 19-9). METHODS This retrospective study included 26 and 39 patients with pathologically confirmed non-hypervascular PNENs and CA 19-9-negative PDACs, respectively, who underwent contrast-enhanced DECT before treatment between June 2019 and December 2021. The clinical, conventional CT qualitative, conventional CT quantitative, and DECT quantitative parameters of the two groups were compared using univariate analysis and selected by least absolute shrinkage and selection operator regression (LASSO) analysis. Multivariate logistic regression analyses were performed to build qualitative, conventional CT quantitative, DECT quantitative, and comprehensive models. The areas under the receiver operating characteristic curve (AUCs) of the models were compared using DeLong's test. RESULTS The AUCs of the DECT quantitative (based on normalized iodine concentrations [nICs] in the arterial and portal venous phases: 0.918; 95% confidence interval [CI] 0.852-0.985) and comprehensive (based on tumour location and nICs in the arterial and portal venous phases: 0.966; 95% CI 0.889-0.995) models were higher than those of the qualitative (based on tumour location: 0.782; 95% CI 0.665-0.899) and conventional CT quantitative (based on normalized conventional CT attenuation in the arterial phase: 0.665; 95% CI 0.533-0.797; all P < 0.05) models. The DECT quantitative and comprehensive models had comparable performances (P = 0.076). CONCLUSIONS Higher nICs in the arterial and portal venous phases were associated with higher blood supply improving the identification of non-hypervascular PNENs.
Collapse
Affiliation(s)
- Xuefang Hu
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000, Guangdong, China
| | - Siya Shi
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Yangdi Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Jiaxin Yuan
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Mingjie Chen
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Luyong Wei
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Weiwei Deng
- Clinical and Technical Support, Philips Healthcare China, Shanghai, 200072, China
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Zhenpeng Peng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China.
| | - Yanji Luo
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
18
|
Winslow ER. Invited Commentary: Symbiotic Relationship between the Pancreatic Surgeon and the Pancreatic Imager: Necessity of Reciprocal Communication. Radiographics 2024; 44:e230225. [PMID: 38060423 DOI: 10.1148/rg.230225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Affiliation(s)
- Emily R Winslow
- From the Department of Surgery, University of Wisconsin Hospitals and Clinics, 700 Highland Ave, Madison, WI 53705
| |
Collapse
|
19
|
Hopstaken JS, Daamen LA, Patijn GA, de Vos-Geelen J, Festen S, Bonsing BA, Verheij M, Hermans JJ, Bruno MJ, de Wilde RF, de Hingh IHJT, Besselink MG, Laarhoven KJHMV, Stommel MWJ. Nationwide evaluation of pancreatic cancer networks ten years after the centralization of pancreatic surgery. HPB (Oxford) 2023; 25:1513-1522. [PMID: 37580180 DOI: 10.1016/j.hpb.2023.07.904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/26/2023] [Accepted: 07/24/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND Due to centralization of pancreatic surgery, patients with pancreatic cancer are treated in pancreatic cancer networks, composed of referring hospitals (Spokes) and an expert center (Hub). This study aimed to investigate I) how pancreatic cancer networks are organized and II) evaluated by involved clinicians. METHODS Two online surveys were sent out between January-May 2022. Part I was sent out to the surgical network directors of all hospitals of the Dutch Pancreatic Cancer Group (DPCG). Part II was sent out to all involved clinicians in the Hubs-and-Spokes networks. RESULTS There was a large variety between the 15 networks concerning number of affiliated Spokes (1-7), annual pancreatoduodenectomies (20-129), and use of a service level agreement (SLA) (40%). More Spoke clinicians considered the Spoke the best location for diagnostic workup (74% vs 36%, P < 0.001). Only 30% of Spoke clinicians attended the Hubs multidisciplinary team meeting frequently. More Hub clinicians thought that exchange of patient information should be improved (37% vs 51%, P = 0.005). CONCLUSION A large variety in Dutch pancreatic cancer networks was observed concerning number of affiliated Spokes, use of SLAs, and logistic aspects of network care. Improvement of network care concern agreements on diagnostic workup, use of SLA, Spoke participation in the MDT, and patient information exchange.
Collapse
Affiliation(s)
| | - Lois A Daamen
- Department of Surgery, UMC Utrecht Cancer Center, Utrecht, the Netherlands; Dept. of Radiation Oncology, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| | - Gijs A Patijn
- Department of Surgery, Isala Oncology Center, Zwolle, the Netherlands
| | - Judith de Vos-Geelen
- Department of Internal Medicine, Division of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht UMC+, Maastricht, the Netherlands
| | | | | | - Marcel Verheij
- Department of Radiation Oncology, Radboudumc, Nijmegen, the Netherlands
| | - John J Hermans
- Department of Medical Imaging, Radboudumc, Nijmegen, the Netherlands
| | - Marco J Bruno
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Roeland F de Wilde
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | | | - Marc G Besselink
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | | | | |
Collapse
|
20
|
Li B, Ni J, Chen F, Lu F, Zhang L, Wu W, Zhang Z. Evaluation of three-dimensional dual-energy CT cholangiopancreatography image quality in patients with pancreatobiliary dilatation: Comparison with conventional single-energy CT. Eur J Radiol Open 2023; 11:100537. [PMID: 37942123 PMCID: PMC10628547 DOI: 10.1016/j.ejro.2023.100537] [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: 07/14/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023] Open
Abstract
Objective This study aimed to evaluate three-dimensional (3D) negative-contrast CT cholangiopancreatography (nCTCP) image quality using dual-energy CT (DECT) with iterative reconstruction (IR) technique in patients with pancreatobiliary dilatation compared with single-energy CT (SECT). Methods Of the patients, 67 and 56 underwent conventional SECT (SECT set) and DECT with IR technique (DECT set), respectively. All patients were retrospectively analyzed during the portal phase to compare objective image quality and other data including patient demographics, hepatic and pancreatic parenchymal enhancement, noise, and attenuation difference (AD) between dilated ducts and enhanced hepatic parenchyma, signal-to-noise ratio (SNR), contrast-to-noise ratio (CNR), and CT volume dose index (CTDIvol). Two radiologists used the five-point Likert scale to evaluate the subjective image quality of 3D nCTCP regarding image noise, sharpness of dilated ducts, and overall image quality. Statistical analyses used the Mann-Whitney U test. Results No significant difference in patient demographics in either CT set was showed during objective evaluation (p > 0.05). However, higher hepatic and pancreatic parenchymal enhancement, AD, SNR, and CNR and lower hepatic and pancreatic noise (p < 0.005) as well as CTDIvol (p = 0.005) on DECT than on SECT were observed. Higher mean grades on DECT than on SECT were showed for image noise (4.65 vs 3.92), sharpness of dilated ducts (4.52 vs 3.94), and overall image quality (4.45 vs 3.91; p < 0.001), respectively during subjective evaluation. Conclusion A higher overall image quality and lower radiation dose on 3D nCTCP can be obtained by DECT with IR technique than with conventional SECT in patients with pancreatobiliary dilatation.
Collapse
Affiliation(s)
- Bin Li
- Department of Radiology, Wuxi No.2 People’s Hospital, 68 Zhong shan Rd., Wuxi 214002, Jiangsu, PR China
| | - JianMing Ni
- Department of Radiology, Wuxi No.2 People’s Hospital, 68 Zhong shan Rd., Wuxi 214002, Jiangsu, PR China
| | - FangMing Chen
- Department of Radiology, Wuxi No.2 People’s Hospital, 68 Zhong shan Rd., Wuxi 214002, Jiangsu, PR China
| | - FengQi Lu
- Department of Radiology, Wuxi No.2 People’s Hospital, 68 Zhong shan Rd., Wuxi 214002, Jiangsu, PR China
| | - Lei Zhang
- Department of Radiology, Wuxi No.2 People’s Hospital, 68 Zhong shan Rd., Wuxi 214002, Jiangsu, PR China
| | - WenJuan Wu
- Department of Radiology, Wuxi No.2 People’s Hospital, 68 Zhong shan Rd., Wuxi 214002, Jiangsu, PR China
| | - ZhuiYang Zhang
- Department of Radiology, Wuxi No.2 People’s Hospital, 68 Zhong shan Rd., Wuxi 214002, Jiangsu, PR China
| |
Collapse
|
21
|
Pacella G, Brunese MC, D’Imperio E, Rotondo M, Scacchi A, Carbone M, Guerra G. Pancreatic Ductal Adenocarcinoma: Update of CT-Based Radiomics Applications in the Pre-Surgical Prediction of the Risk of Post-Operative Fistula, Resectability Status and Prognosis. J Clin Med 2023; 12:7380. [PMID: 38068432 PMCID: PMC10707069 DOI: 10.3390/jcm12237380] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 09/10/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is the seventh leading cause of cancer-related deaths worldwide. Surgical resection is the main driver to improving survival in resectable tumors, while neoadjuvant treatment based on chemotherapy (and radiotherapy) is the best option-treatment for a non-primally resectable disease. CT-based imaging has a central role in detecting, staging, and managing PDAC. As several authors have proposed radiomics for risk stratification in patients undergoing surgery for PADC, in this narrative review, we have explored the actual fields of interest of radiomics tools in PDAC built on pre-surgical imaging and clinical variables, to obtain more objective and reliable predictors. METHODS The PubMed database was searched for papers published in the English language no earlier than January 2018. RESULTS We found 301 studies, and 11 satisfied our research criteria. Of those included, four were on resectability status prediction, three on preoperative pancreatic fistula (POPF) prediction, and four on survival prediction. Most of the studies were retrospective. CONCLUSIONS It is possible to conclude that many performing models have been developed to get predictive information in pre-surgical evaluation. However, all the studies were retrospective, lacking further external validation in prospective and multicentric cohorts. Furthermore, the radiomics models and the expression of results should be standardized and automatized to be applicable in clinical practice.
Collapse
Affiliation(s)
- Giulia Pacella
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (G.P.)
| | - Maria Chiara Brunese
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (G.P.)
| | | | - Marco Rotondo
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (G.P.)
| | - Andrea Scacchi
- General Surgery Unit, University of Milano-Bicocca, 20126 Milan, Italy
| | - Mattia Carbone
- San Giovanni di Dio e Ruggi d’Aragona Hospital, 84131 Salerno, Italy;
| | - Germano Guerra
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (G.P.)
| |
Collapse
|
22
|
Conroy T, Pfeiffer P, Vilgrain V, Lamarca A, Seufferlein T, O'Reilly EM, Hackert T, Golan T, Prager G, Haustermans K, Vogel A, Ducreux M. Pancreatic cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2023; 34:987-1002. [PMID: 37678671 DOI: 10.1016/j.annonc.2023.08.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Affiliation(s)
- T Conroy
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Vandoeuvre-lès-Nancy; APEMAC, équipe MICS, Université de Lorraine, Nancy, France
| | - P Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - V Vilgrain
- Centre de Recherche sur l'Inflammation U 1149, Université Paris Cité, Paris; Department of Radiology, Beaujon Hospital, APHP Nord, Clichy, France
| | - A Lamarca
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - T Seufferlein
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - E M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - T Hackert
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - T Golan
- Gastrointestinal Unit, Oncology Institute, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - G Prager
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - K Haustermans
- Department of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - A Vogel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - M Ducreux
- Université Paris-Saclay, Gustave Roussy, Inserm Unité Dynamique des Cellules Tumorales, Villejuif, France
| |
Collapse
|
23
|
Lim DH, Yoon H, Kim KP, Ryoo BY, Lee SS, Park DH, Song TJ, Hwang DW, Lee JH, Song KB, Kim SC, Hong SM, Hyung J, Yoo C. Analysis of Plasma Circulating Tumor DNA in Borderline Resectable Pancreatic Cancer Treated with Neoadjuvant Modified FOLFIRINOX: Clinical Relevance of DNA Damage Repair Gene Alteration Detection. Cancer Res Treat 2023; 55:1313-1320. [PMID: 37139665 PMCID: PMC10582539 DOI: 10.4143/crt.2023.452] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/04/2023] [Indexed: 05/05/2023] Open
Abstract
PURPOSE There are no reliable biomarkers to guide treatment for patients with borderline resectable pancreatic cancer (BRPC) in the neoadjuvant setting. We used plasma circulating tumor DNA (ctDNA) sequencing to search biomarkers for patients with BRPC receiving neoadjuvant mFOLFIRINOX in our phase 2 clinical trial (NCT02749136). MATERIALS AND METHODS Among the 44 patients enrolled in the trial, patients with plasma ctDNA sequencing at baseline or post-operation were included in this analysis. Plasma cell-free DNA isolation and sequencing were performed using the Guardant 360 assay. Detection of genomic alterations, including DNA damage repair (DDR) genes, were examined for correlations with survival. RESULTS Among the 44 patients, 28 patients had ctDNA sequencing data qualified for the analysis and were included in this study. Among the 25 patients with baseline plasma ctDNA data, 10 patients (40%) had alterations of DDR genes detected at baseline, inclu-ding ATM, BRCA1, BRCA2 and MLH1, and showed significantly better progression-free survival than those without such DDR gene alterations detected (median, 26.6 vs. 13.5 months; log-rank p=0.004). Patients with somatic KRAS mutations detected at baseline (n=6) had significantly worse overall survival (median, 8.5 months vs. not applicable; log-rank p=0.003) than those without. Among 13 patients with post-operative plasma ctDNA data, eight patients (61.5%) had detectable somatic alterations. CONCLUSION Detection of DDR gene mutations from plasma ctDNA at baseline was associated with better survival outcomes of pati-ents with borderline resectable pancreatic ductal adenocarcinoma treated with neoadjuvant mFOLFIRINOX and may be a prognostic biomarker.
Collapse
Affiliation(s)
- Dong-Hoon Lim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Hyunseok Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Kyu-pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Baek-Yeol Ryoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Sang Soo Lee
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Do Hyun Park
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Tae Jun Song
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Dae Wook Hwang
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Jae Hoon Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Ki Byung Song
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Song Cheol Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Jaewon Hyung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| |
Collapse
|
24
|
Yang C, Guo J, Chen L, Zhang L, Zhang L, Li K, Chen J, Chen W. The dose of focused ultrasound ablation surgery (FUAS) for unresectable pancreatic cancer is predictable: A multicenter retrospective study. Medicine (Baltimore) 2023; 102:e34684. [PMID: 37746965 PMCID: PMC10519495 DOI: 10.1097/md.0000000000034684] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 09/26/2023] Open
Abstract
To analyze the influencing factors of energy efficiency factors (EEF) in focused ultrasound ablation surgery (FUAS) for unresectable pancreatic cancer and build a dosimetry model. The patients with unresectable pancreatic cancer that underwent FUAS were enrolled from 3 clinical centers between June 2015 and June 2022 for retrospective analysis. The significance of the factors with the potential to affect the EEF was assessed, correlations among the factors were analyzed, and the accuracy of the prediction models established by the factors containing different imaging features was compared. From a total of 236 cases, 215 cases were screened for study, EEF was significantly correlated with mode of anesthesia, grayscale change, tumor volume, tumor location, the distance from the tumor center to skin, contrast-enhanced computer tomography enhancement type, T2-weighted imaging fat suppression signal intensity and contrast-enhanced T1-weighted imaging enhancement type on magnetic resonance imaging. The resultant multiple regression models of EEF achieved significance, contains predictors of Tumor volume, the distance from tumor center to skin, T2-weighted imaging fat suppression signal intensity, and contrast-enhanced T1-weighted imaging enhancement type had better goodness of fit. Compared with CT, the EEF prediction model established by adding magnetic resonance imaging features showed better prediction in FUAS treatment of unresectable pancreatic cancer.
Collapse
Affiliation(s)
- Chao Yang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Jing Guo
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, P.R. China
| | - Li Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Department of Oncology, Chongqing Haifu Hospital, Chongqing, P.R. China
| | - Luping Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Lian Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Department of Oncology, Chongqing Haifu Hospital, Chongqing, P.R. China
| | - Kequan Li
- Department of Oncology, Chongqing Haifu Hospital, Chongqing, P.R. China
| | - Jinyun Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Wenzhi Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
25
|
Roesel R, Deantonio L, Bernardi L, Garo ML, Majno-Hurst P, Vannelli A, Cefalì M, Palmarocchi MC, Valli MC, Pesola G, Cristaudi A, De Dosso S. Neo-Adjuvant Treatment in Primary Resectable Pancreatic Cancer: A Systematic Review and PRISMA-Compliant Updated Metanalysis of Oncological Outcomes. Cancers (Basel) 2023; 15:4627. [PMID: 37760596 PMCID: PMC10526896 DOI: 10.3390/cancers15184627] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Despite advances in treatment, the prognosis of resectable pancreatic adenocarcinoma remains poor. Neoadjuvant therapy (NAT) has gained great interest in hopes of improving survival. However, the results of available studies based on different treatment approaches, such as chemotherapy and chemoradiotherapy, showed contrasting results. The aim of this systematic review and meta-analysis is to clarify the benefit of NAT compared to upfront surgery (US) in primarily resectable pancreatic adenocarcinoma. METHODS A PRISMA literature review identified 139 studies, of which 15 were finally included in the systematic review and meta-analysis. All data from eligible articles was summarized in a systematic summary and then used for the meta-analysis. Specifically, we used HR for OS and DFS and risk estimates (odds ratios) for the R0 resection rate and the N+ rate. The risk of bias was correctly assessed according to the nature of the studies included. RESULTS From the pooled HRs, OS for NAT patients was better, with an HR for death of 0.80 (95% CI: 0.72-0.90) at a significance level of less than 1%. In the sub-group analysis, no difference was found between patients treated with chemoradiotherapy or chemotherapy exclusively. The meta-analysis of seven studies that reported DFS for NAT resulted in a pooled HR for progression of 0.66 (95% CI: 0.56-0.79) with a significance level of less than 1%. A significantly lower risk of positive lymph nodes (OR: 0.45; 95% CI: 0.32-0.63) and an improved R0 resection rate (OR: 1.70; 95% CI: 1.23-2.36) were also found in patients treated with NAT, despite high heterogeneity. CONCLUSIONS NAT is associated with improved survival for patients with resectable pancreatic adenocarcinoma; however, the optimal treatment strategy has yet to be defined, and further studies are required.
Collapse
Affiliation(s)
- Raffaello Roesel
- Department of Visceral Surgery, Hospital of Lugano (EOC), 6900 Lugano, Switzerland (P.M.-H.); (A.C.)
| | - Letizia Deantonio
- Radiation Oncology Department, Oncology Institute of Southern Switzerland (IOSI), EOC, 6500 Bellinzona, Switzerland; (L.D.); (M.C.V.)
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), 6900 Lugano, Switzerland
| | - Lorenzo Bernardi
- Department of Visceral Surgery, Hospital of Lugano (EOC), 6900 Lugano, Switzerland (P.M.-H.); (A.C.)
| | | | - Pietro Majno-Hurst
- Department of Visceral Surgery, Hospital of Lugano (EOC), 6900 Lugano, Switzerland (P.M.-H.); (A.C.)
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), 6900 Lugano, Switzerland
| | - Alberto Vannelli
- Department of General Surgery, Ospedale Valduce, 22100 Como, Italy;
| | - Marco Cefalì
- Medical Oncology Department, Oncology Institute of Southern Switzerland (IOSI), EOC, 6500 Bellinzona, Switzerland; (M.C.); (M.C.P.); (G.P.)
| | - Maria Celeste Palmarocchi
- Medical Oncology Department, Oncology Institute of Southern Switzerland (IOSI), EOC, 6500 Bellinzona, Switzerland; (M.C.); (M.C.P.); (G.P.)
| | - Maria Carla Valli
- Radiation Oncology Department, Oncology Institute of Southern Switzerland (IOSI), EOC, 6500 Bellinzona, Switzerland; (L.D.); (M.C.V.)
| | - Guido Pesola
- Medical Oncology Department, Oncology Institute of Southern Switzerland (IOSI), EOC, 6500 Bellinzona, Switzerland; (M.C.); (M.C.P.); (G.P.)
| | - Alessandra Cristaudi
- Department of Visceral Surgery, Hospital of Lugano (EOC), 6900 Lugano, Switzerland (P.M.-H.); (A.C.)
| | - Sara De Dosso
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), 6900 Lugano, Switzerland
- Medical Oncology Department, Oncology Institute of Southern Switzerland (IOSI), EOC, 6500 Bellinzona, Switzerland; (M.C.); (M.C.P.); (G.P.)
| |
Collapse
|
26
|
Ueda H, Takahashi H, Kobayashi S, Sasaki K, Iwagami Y, Yamada D, Tomimaru Y, Asaoka T, Noda T, Tanemura M, Doki Y, Eguchi H. Pancreatic cancer near the splenic hilum has a higher likelihood of splenic vessel invasion and unfavorable survival. Langenbecks Arch Surg 2023; 408:353. [PMID: 37695403 DOI: 10.1007/s00423-023-03089-8] [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: 06/19/2023] [Accepted: 08/29/2023] [Indexed: 09/12/2023]
Abstract
PURPOSE This study aimed to investigate whether clinical outcomes varied based on the tumor location within the pancreatic body and tail in patients with pancreatic cancer (PC). METHODS Ninety-five patients who had undergone a distal pancreatectomy for resectable (R) or borderline resectable (BR) PC within the pancreatic body or tail region were retrospectively investigated and divided into four groups (three subgroups of R-PC according to tumor location, and BR-PC): R-PC in the pancreatic body region (group A, n = 24), R-PC on the right side of the pancreatic tail region (group B, n = 17), R-PC on the left side of the pancreatic tail region (group C, n = 29), and BR-PC located in any region within the pancreatic body and tail (group BR, n = 25). RESULTS Group C patients showed a higher incidence of pretreatment splenic artery and vein involvement than group A and B patients (splenic artery: 8.3/11.8/41.4%, p < 0.010; splenic vein: 25.0/23.5/79.3%, p < 0.010, in groups A/B/C, respectively). The overall survival of group C patients was significantly unfavorable compared to that of group A and B patients (median: 3.9/4.2/2.3 years in groups A/B/C, p = 0.029, respectively). Pretreatment clinical factors were comparable between group C and group BR. Median survival rates were comparable between group C and BR patients (2.3 and 2.0 years, respectively) (p = 0.93). CONCLUSIONS Differences in anatomical location within the pancreatic body and tail characterize the unfavorable outcomes of PC near the splenic hilum.
Collapse
Affiliation(s)
- Hiroki Ueda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hidenori Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kazuki Sasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshihumi Iwagami
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Daisaku Yamada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshito Tomimaru
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tadafumi Asaoka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Surgery, Osaka Police Hospital, Osaka, Japan
| | - Takehiro Noda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masahiro Tanemura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Surgery, Rinku General Medical Center, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
27
|
Gieser P, Merle U, Junghanss T, Weber TF, Stojković M. Vascular pathology in patients with alveolar echinococcosis: framework for assessment and clinical management - a retrospective case series. BMJ Open Gastroenterol 2023; 10:e001181. [PMID: 37567730 PMCID: PMC10423801 DOI: 10.1136/bmjgast-2023-001181] [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: 04/24/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
OBJECTIVE Alveolar echinococcosis (AE) is a parasitic liver disease with infiltrative growth similar to solid organ malignancies. Major vascular damage is frequent and often remains untreated until catastrophic events precipitate. Detailed clinical and radiological assessment is required to guide individualised treatment decisions. Standardised radiological reporting templates of malignancies with profiles resembling AE are candidates for adaptation. Our objectives are to describe vascular pathology in AE and establish a framework for structured evaluation as the basis for treatment decisions and monitoring. DESIGN Retrospective case series. RESULTS 69 patients (37.1%) had vascular involvement: portal vein (PV) 24.7%, hepatic vein (HV) 22.6% inferior vena cava (IVC) 13.4%. Significant stenosis/occlusion of vessels was present in 15.1% of PV, in 13.4% of HV and in 7.5% of IVC involvement. Vascular pathology needing specific treatment or monitoring was present in 8.6% of patients. The most frequent clinical presentation was high grade IVC stenosis or occlusion which was seen in 11 patients of the cohort. CONCLUSION Advanced AE requires early multidisciplinary assessment to prevent progressive impairment of liver function due to vascular damage. The focus at first presentation is on complete evaluation of vascular (and biliary) involvement. The focus in non-resectable AE is on prevention of vascular (and biliary) complications while suppressing growth of AE lesions by benzimidazole treatment to improve the quality of life of patients. We developed a framework for standardised vascular assessment and follow-up of patients with AE to recognise and treat complications early.
Collapse
Affiliation(s)
- Paula Gieser
- Infectious and Tropical Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Uta Merle
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Junghanss
- Infectious and Tropical Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Tim F Weber
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Marija Stojković
- Infectious and Tropical Diseases, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
28
|
Megibow AJ. Pancreatic Cysts: Radiology. Gastrointest Endosc Clin N Am 2023; 33:519-531. [PMID: 37245933 DOI: 10.1016/j.giec.2023.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
This article reviews the types of pancreatic cysts encountered in Radiologic practice. It summarizes the malignancy risk of each of the following: serous cystadenoma, mucinous cystic tumor, intraductal papillary mucinous neoplasm main duct and side branch, and some miscellaneous cysts such as neuroendocrine tumor and solid pseudopapillary epithelial neoplasm. Specific reporting recommendations are given. The choice between radiology follow-up versus endoscopic analysis is discussed.
Collapse
Affiliation(s)
- Alec J Megibow
- Department of Radiology, NYU-Langone Health, 550 1st Avenue, Room HCC 232, New York, NY 10016, USA.
| |
Collapse
|
29
|
Taya M, Hecht EM, Huang C, Lo GC. Pancreatic Cystic Lesions: Imaging Techniques and Diagnostic Features. Gastrointest Endosc Clin N Am 2023; 33:497-518. [PMID: 37245932 DOI: 10.1016/j.giec.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The detection of incidental pancreatic cystic lesions has increased over time. It is crucial to separate benign from potentially malignant or malignant lesions to guide management and reduce morbidity and mortality. The key imaging features used to fully characterize cystic lesions are optimally assessed by contrast-enhanced magnetic resonance imaging/magnetic resonance cholangiopancreatography, with pancreas protocol computed tomography offering a complementary role. While some imaging features have high specificity for a particular diagnosis, overlapping imaging features between diagnoses may require further investigation with follow-up diagnostic imaging or tissue sampling.
Collapse
Affiliation(s)
- Michio Taya
- Department of Radiology, New York Presbyterian - Weill Cornell Medicine, 520 East 70th Street, Starr 8a, New York, NY 10021, USA
| | - Elizabeth M Hecht
- Department of Radiology, New York Presbyterian - Weill Cornell Medicine, 520 East 70th Street, Starr 8a, New York, NY 10021, USA
| | - Chenchan Huang
- Department of Radiology, NYU Grossman School of Medicine, 560 1st Avenue, 2F, New York, NY 10016, USA
| | - Grace C Lo
- Department of Radiology, New York Presbyterian - Weill Cornell Medicine, 520 East 70th Street, Starr 8a, New York, NY 10021, USA.
| |
Collapse
|
30
|
Kimura J, Sui K, Tabuchi M, Sakamoto S, Murokawa T, Iwata J, Morita S, Iiyama T, Okabayashi T. Preoperative percent body fat in bioelectrical impedance analysis predicts pancreatic fistula after pancreaticoduodenectomy. Langenbecks Arch Surg 2023; 408:248. [PMID: 37380749 DOI: 10.1007/s00423-023-02998-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: 10/18/2022] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND It is unclear which body composition affects postoperative pancreatic fistula (POPF) after pancreaticoduodenectomy. In the present study, we evaluated the relationship between nutritional factors, body composition, and POPF. METHODS This was a prospective observational cohort study. Patients who underwent pancreaticoduodenectomy between March 2018 and July 2021 were included in this study. Preoperative body composition was measured using a bioelectrical impedance analyzer. In addition, the predictive factors for POPF were analyzed using logistic regression model. RESULTS The study included 143 patients. Among these patients, 31 had POPF (POPF group) and 112 did not (non-POPF group) after pancreaticoduodenectomy. For body composition, the percent body fat was significantly higher in the POPF group (26.90 vs 23.48, P = 0.022). Multivariate analysis revealed that alcohol consumption (odds ratio 2.95, P = 0.03), pancreatic duct < 3 mm (odds ratio 3.89, P < 0.01), and percent body fat (odds ratio 1.08, P = 0.01) were significantly independent predictive factors for POPF. When the patients were divided into three groups based on their percent body fat (< 25, 25-35, and ≥ 35), POPF occurred more frequently in the group with ≥ 35 percent body fat (47.1%) than in the < 25 group (15.5%) (P = 0.008). CONCLUSION Predictive factors for POPF related to nutritional status, such as percent body fat, should be considered before proceeding to pancreaticoduodenectomy (ClinicalTrials.gov trial registration no. NCT5257434).
Collapse
Affiliation(s)
- Jiro Kimura
- Department of Gastroenterological Surgery, Kochi Health Sciences Center, 2125-1, Ike, Kochi City, Kochi, 781-8555, Japan
| | - Kenta Sui
- Department of Gastroenterological Surgery, Kochi Health Sciences Center, 2125-1, Ike, Kochi City, Kochi, 781-8555, Japan
| | - Motoyasu Tabuchi
- Department of Gastroenterological Surgery, Kochi Health Sciences Center, 2125-1, Ike, Kochi City, Kochi, 781-8555, Japan
| | - Shinya Sakamoto
- Department of Gastroenterological Surgery, Kochi Health Sciences Center, 2125-1, Ike, Kochi City, Kochi, 781-8555, Japan
| | - Takahiro Murokawa
- Department of Gastroenterological Surgery, Kochi Health Sciences Center, 2125-1, Ike, Kochi City, Kochi, 781-8555, Japan
| | - Jun Iwata
- Department of Diagnostic Pathology, Kochi Health Sciences Center, Kochi City, Kochi, Japan
| | - Sojiro Morita
- Department of Radiology, Kochi Health Sciences Center, Kochi City, Kochi, Japan
| | - Tastuo Iiyama
- Department of Biostatistics, National Center for Global Health and Medicine, Shinjuku City, Japan
| | - Takehiro Okabayashi
- Department of Gastroenterological Surgery, Kochi Health Sciences Center, 2125-1, Ike, Kochi City, Kochi, 781-8555, Japan.
| |
Collapse
|
31
|
Ramaekers M, Viviers CGA, Janssen BV, Hellström TAE, Ewals L, van der Wulp K, Nederend J, Jacobs I, Pluyter JR, Mavroeidis D, van der Sommen F, Besselink MG, Luyer MDP. Computer-Aided Detection for Pancreatic Cancer Diagnosis: Radiological Challenges and Future Directions. J Clin Med 2023; 12:4209. [PMID: 37445243 DOI: 10.3390/jcm12134209] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Radiological imaging plays a crucial role in the detection and treatment of pancreatic ductal adenocarcinoma (PDAC). However, there are several challenges associated with the use of these techniques in daily clinical practice. Determination of the presence or absence of cancer using radiological imaging is difficult and requires specific expertise, especially after neoadjuvant therapy. Early detection and characterization of tumors would potentially increase the number of patients who are eligible for curative treatment. Over the last decades, artificial intelligence (AI)-based computer-aided detection (CAD) has rapidly evolved as a means for improving the radiological detection of cancer and the assessment of the extent of disease. Although the results of AI applications seem promising, widespread adoption in clinical practice has not taken place. This narrative review provides an overview of current radiological CAD systems in pancreatic cancer, highlights challenges that are pertinent to clinical practice, and discusses potential solutions for these challenges.
Collapse
Affiliation(s)
- Mark Ramaekers
- Department of Surgery, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands
| | - Christiaan G A Viviers
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Boris V Janssen
- Department of Surgery, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Terese A E Hellström
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Lotte Ewals
- Department of Radiology, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands
| | - Kasper van der Wulp
- Department of Radiology, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands
| | - Joost Nederend
- Department of Radiology, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands
| | - Igor Jacobs
- Department of Hospital Services and Informatics, Philips Research, 5656 AE Eindhoven, The Netherlands
| | - Jon R Pluyter
- Department of Experience Design, Philips Design, 5656 AE Eindhoven, The Netherlands
| | - Dimitrios Mavroeidis
- Department of Data Science, Philips Research, 5656 AE Eindhoven, The Netherlands
| | - Fons van der Sommen
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Marc G Besselink
- Department of Surgery, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Misha D P Luyer
- Department of Surgery, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands
| |
Collapse
|
32
|
Fiore M, Coppola A, Petrianni GM, Trecca P, D’Ercole G, Cimini P, Ippolito E, Caputo D, Beomonte Zobel B, Coppola R, Ramella S. Advances in pre-treatment evaluation of pancreatic ductal adenocarcinoma: a narrative review. J Gastrointest Oncol 2023; 14:1114-1130. [PMID: 37201095 PMCID: PMC10186502 DOI: 10.21037/jgo-22-1034] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/08/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Despite advances in the multidisciplinary management of pancreatic cancer, overall prognosis remains poor, due to early progression of the disease. There is a need to also take action in staging, to make it increasingly accurate and complete, to define the setting of the therapeutic strategy. This review was planned to update the current status of pre-treatment evaluation for pancreatic cancer. METHODS We conducted an extensive review, including relevant articles dealing with traditional imaging, functional imaging and minimally invasive surgical procedures before treatment for pancreatic cancer. We searched articles written in English only. Data in the PubMed database, published in the period between January 2000 and January 2022, were retrieved. Prospective observational studies, retrospective analyses and meta-analyses were reviewed and analysed. KEY CONTENT AND FINDINGS Each imaging modality (endoscopic ultrasonography, endoscopic retrograde cholangiopancreatography, computed tomography, positron emission tomography/computed tomography, staging laparoscopy) has its own diagnostic advantages and limitations. The sensitivity, specificity and accuracy for each image set are reported. Data that support the increasing role of neoadjuvant therapy (radiotherapy and chemotherapy) and the meaning of a patient-tailored treatment selection, based on tumour staging, are also discussed. CONCLUSIONS A multimodal pre-treatment workup should be searched as it improves staging accuracy, orienting patients with resectable tumors towards surgery, optimizing patient selection with locally advanced tumors to neoadjuvant or definite therapy and avoiding surgical resection or curative radiotherapy in those with metastatic disease.
Collapse
Affiliation(s)
- Michele Fiore
- Research Unit of Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | | | - Gian Marco Petrianni
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Pasquale Trecca
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Gabriele D’Ercole
- Research Unit of Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Paola Cimini
- Operative Research Unit of Radiology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Edy Ippolito
- Research Unit of Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Damiano Caputo
- Department of Surgery and Research Unit of General Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Research Unit of General Surgery Unit Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Bruno Beomonte Zobel
- Operative Research Unit of Radiology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Roberto Coppola
- Department of Surgery and Research Unit of General Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Research Unit of General Surgery Unit Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Sara Ramella
- Research Unit of Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| |
Collapse
|
33
|
Gu Q, He M, He Y, Dai A, Liu J, Chen X, Liu P. CT-measured body composition radiomics predict lymph node metastasis in localized pancreatic ductal adenocarcinoma. Discov Oncol 2023; 14:16. [PMID: 36735166 PMCID: PMC9898483 DOI: 10.1007/s12672-023-00624-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/31/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND To explored the value of CT-measured body composition radiomics in preoperative evaluation of lymph node metastasis (LNM) in localized pancreatic ductal adenocarcinoma (LPDAC). METHODS We retrospectively collected patients with LPDAC who underwent surgical resection from January 2016 to June 2022. According to whether there was LNM after operation, the patients were divided into LNM group and non-LNM group in both male and female patients. The patient's body composition was measured by CT images at the level of the L3 vertebral body before surgery, and the radiomics features of adipose tissue and muscle were extracted. Multivariate logistic regression (forward LR) analyses were used to determine the predictors of LNM from male and female patient, respectively. Sexual dimorphism prediction signature using adipose tissue radiomics features, muscle tissue radiomics features and combined signature of both were developed and compared. The model performance is evaluated on discrimination and validated through a leave-one-out cross-validation method. RESULTS A total of 196 patients (mean age, 60 years ± 9 [SD]; 117 men) were enrolled, including 59 LNM in male and 36 LNM in female. Both male and female CT-measured body composition radiomics signatures have a certain predictive power on LNM of LPDAC. Among them, the female adipose tissue signature showed the highest performance (area under the ROC curve (AUC), 0.895), and leave one out cross validation (LOOCV) indicated that the signature could accurately classify 83.5% of cases; The prediction efficiency of the signature can be further improved after adding the muscle radiomics features (AUC, 0.924, and the accuracy of the LOOCV was 87.3%); The abilities of male adipose tissue and muscle tissue radiomics signatures in predicting LNM of LPDAC was similar, AUC was 0.735 and 0.773, respectively, and the accuracy of LOOCV was 62.4% and 68.4%, respectively. CONCLUSIONS CT-measured body composition Radiomics strategy showed good performance for predicting LNM in LPDAC, and has sexual dimorphism. It may provide a reference for individual treatment of LPDAC and related research about body composition in the future.
Collapse
Affiliation(s)
- Qianbiao Gu
- Department of Radiology, Hunan Provincial People’s Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005 China
| | - Mengqing He
- Department of Radiology, Hunan Provincial People’s Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005 China
| | - Yaqiong He
- Department of Radiology, Hunan Provincial People’s Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005 China
| | - Anqi Dai
- Department of Radiology, Hunan Provincial People’s Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005 China
| | - Jianbin Liu
- Department of Radiology, Hunan Provincial People’s Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005 China
| | - Xiang Chen
- Department of Radiology, Hunan Provincial People’s Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005 China
| | - Peng Liu
- Department of Radiology, Hunan Provincial People’s Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005 China
| |
Collapse
|
34
|
Li Q, Li X, Liu W, Yu J, Chen Y, Zhu M, Li N, Liu F, Wang T, Fang X, Li J, Lu J, Shao C, Bian Y. Non-enhanced magnetic resonance imaging-based radiomics model for the differentiation of pancreatic adenosquamous carcinoma from pancreatic ductal adenocarcinoma. Front Oncol 2023; 13:1108545. [PMID: 36756153 PMCID: PMC9900003 DOI: 10.3389/fonc.2023.1108545] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023] Open
Abstract
Purpose To evaluate the diagnostic performance of radiomics model based on fully automatic segmentation of pancreatic tumors from non-enhanced magnetic resonance imaging (MRI) for differentiating pancreatic adenosquamous carcinoma (PASC) from pancreatic ductal adenocarcinoma (PDAC). Materials and methods In this retrospective study, patients with surgically resected histopathologically confirmed PASC and PDAC who underwent MRI scans between January 2011 and December 2020 were included in the study. Multivariable logistic regression analysis was conducted to develop a clinical and radiomics model based on non-enhanced T1-weighted and T2-weighted images. The model performances were determined based on their discrimination and clinical utility. Kaplan-Meier and log-rank tests were used for survival analysis. Results A total of 510 consecutive patients including 387 patients (age: 61 ± 9 years; range: 28-86 years; 250 males) with PDAC and 123 patients (age: 62 ± 10 years; range: 36-84 years; 78 males) with PASC were included in the study. All patients were split into training (n=382) and validation (n=128) sets according to time. The radiomics model showed good discrimination in the validation (AUC, 0.87) set and outperformed the MRI model (validation set AUC, 0.80) and the ring-enhancement (validation set AUC, 0.74). Conclusions The radiomics model based on non-enhanced MRI outperformed the MRI model and ring-enhancement to differentiate PASC from PDAC; it can, thus, provide important information for decision-making towards precise management and treatment of PASC.
Collapse
Affiliation(s)
- Qi Li
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China,Department of Radiology, 96601 Military Hospital of PLA, Huangshan, Anhui, China
| | - Xuezhou Li
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Wenbin Liu
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Jieyu Yu
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Yukun Chen
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Mengmeng Zhu
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Na Li
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Fang Liu
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Tiegong Wang
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Xu Fang
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Jing Li
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Jianping Lu
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Chengwei Shao
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China,*Correspondence: Yun Bian, ; Chengwei Shao,
| | - Yun Bian
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China,*Correspondence: Yun Bian, ; Chengwei Shao,
| |
Collapse
|
35
|
Shukla HD, Dukic T, Roy S, Bhandary B, Gerry A, Poirier Y, Lamichhane N, Molitoris J, Carrier F, Banerjee A, Regine WF, Polf JC. Pancreatic cancer derived 3D organoids as a clinical tool to evaluate the treatment response. Front Oncol 2023; 12:1072774. [PMID: 36713532 PMCID: PMC9879007 DOI: 10.3389/fonc.2022.1072774] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/29/2022] [Indexed: 01/13/2023] Open
Abstract
Background and purpose Pancreatic cancer (PC) is the fourth leading cause of cancer death in both men and women. The standard of care for patients with locally advanced PC of chemotherapy, stereotactic radiotherapy (RT), or chemo-radiation-therapy has shown highly variable and limited success rates. However, three-dimensional (3D) Pancreatic tumor organoids (PTOs) have shown promise to study tumor response to drugs, and emerging treatments under in vitro conditions. We investigated the potential for using 3D organoids to evaluate the precise radiation and drug dose responses of in vivo PC tumors. Methods PTOs were created from mouse pancreatic tumor tissues, and their microenvironment was compared to that of in vivo tumors using immunohistochemical and immunofluorescence staining. The organoids and in vivo PC tumors were treated with fractionated X-ray RT, 3-bromopyruvate (3BP) anti-tumor drug, and combination of 3BP + fractionated RT. Results Pancreatic tumor organoids (PTOs) exhibited a similar fibrotic microenvironment and molecular response (as seen by apoptosis biomarker expression) as in vivo tumors. Untreated tumor organoids and in vivo tumor both exhibited proliferative growth of 6 folds the original size after 10 days, whereas no growth was seen for organoids and in vivo tumors treated with 8 (Gray) Gy of fractionated RT. Tumor organoids showed reduced growth rates of 3.2x and 1.8x when treated with 4 and 6 Gy fractionated RT, respectively. Interestingly, combination of 100 µM of 3BP + 4 Gy of RT showed pronounced growth inhibition as compared to 3-BP alone or 4 Gy of radiation alone. Further, positive identification of SOX2, SOX10 and TGFβ indicated presence of cancer stem cells in tumor organoids which might have some role in resistance to therapies in pancreatic cancer. Conclusions PTOs produced a similar microenvironment and exhibited similar growth characteristics as in vivo tumors following treatment, indicating their potential for predicting in vivo tumor sensitivity and response to RT and combined chemo-RT treatments.
Collapse
Affiliation(s)
- Hem D Shukla
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, United States,*Correspondence: Hem D Shukla,
| | - Tijana Dukic
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Sanjit Roy
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Binny Bhandary
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Andrew Gerry
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Yannick Poirier
- Division of Medical Physics, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Narottam Lamichhane
- Division of Medical Physics, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Jason Molitoris
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - France Carrier
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Aditi Banerjee
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - William F. Regine
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Jerimy C. Polf
- Division of Medical Physics, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, United States
| |
Collapse
|
36
|
Khuntikeo N, Pugkhem A, Srisuk T, Luvira V, Titapun A, Tipwaratorn T, Thanasukarn V, Klungboonkrong V, Wongwiwatchai J. Surgery. Recent Results Cancer Res 2023; 219:147-222. [PMID: 37660334 DOI: 10.1007/978-3-031-35166-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
This chapter provides a comprehensive background from basic to applied knowledge of surgical anatomy which is necessary for the surgical treatment of cholangiocarcinoma (CCA) patients. Significant advances that have been made in the surgical treatment of CCA were examined. For instance, in-depth details are provided for appropriate preoperative assessment and treatment to optimize patient status and to improve the outcome of surgical treatment(s). Comprehensive details are provided for the surgical techniques and outcomes of treatments for each type of CCA with clear illustrations and images. This chapter also describes the role of minimally invasive surgery and liver transplantation in CCA treatment.
Collapse
Affiliation(s)
- Narong Khuntikeo
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand.
| | - Ake Pugkhem
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Tharatip Srisuk
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Vor Luvira
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Attapol Titapun
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Theerawee Tipwaratorn
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Vasin Thanasukarn
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Vivian Klungboonkrong
- Department of Radiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Jitraporn Wongwiwatchai
- Department of Radiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| |
Collapse
|
37
|
Li B, Lu F, Ni J, Wu W, Xu H, Zhang Z. Preoperative evaluation of malignant pancreatobiliary obstruction: A novel technique for multiphase fusion 3D CT images. Eur J Radiol Open 2022; 10:100464. [PMID: 36545431 PMCID: PMC9761364 DOI: 10.1016/j.ejro.2022.100464] [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: 06/16/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
The purpose of this article is to describe a novel technique of multiphase fusion three-dimensional (3D) images in patients with malignant pancreatobiliary obstruction. Multiphase fusion 3D images of CT arteriography, portovenography and hepatic venography combined with negative-contrast CT cholangiopancreatography can be done with enhanced multiphase CT scan using intravenous contrast agent at once. This technique may be feasible for one-stop evaluation of malignant pancreatobiliary obstruction.
Collapse
|
38
|
Effects of Berberine against Pancreatitis and Pancreatic Cancer. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238630. [PMID: 36500723 PMCID: PMC9738201 DOI: 10.3390/molecules27238630] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
The pancreas is a glandular organ with endocrine and exocrine functions necessary for the maintenance of blood glucose homeostasis and secretion of digestive enzymes. Pancreatitis is characterized by inflammation of the pancreas leading to temporary or permanent pancreatic dysfunction. Inflammation and fibrosis caused by chronic pancreatitis exacerbate malignant transformation and significantly increase the risk of developing pancreatic cancer, the world's most aggressive cancer with a 5-year survival rate less than 10%. Berberine (BBR) is a naturally occurring plant-derived polyphenol present in a variety of herbal remedies used in traditional medicine to treat ulcers, infections, jaundice, and inflammation. The current review summarizes the existing in vitro and in vivo evidence on the effects of BBR against pancreatitis and pancreatic cancer with a focus on the signalling mechanisms underlying the effects of BBR.
Collapse
|
39
|
Radiomics Combined with Multiple Machine Learning Algorithms in Differentiating Pancreatic Ductal Adenocarcinoma from Pancreatic Neuroendocrine Tumor: More Hands Produce a Stronger Flame. J Clin Med 2022; 11:jcm11226789. [PMID: 36431266 PMCID: PMC9697420 DOI: 10.3390/jcm11226789] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/04/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
The aim of this study was to assess the diagnostic ability of radiomics combined with multiple machine learning algorithms to differentiate pancreatic ductal adenocarcinoma (PDAC) from pancreatic neuroendocrine tumor (pNET). This retrospective study included a total of 238 patients diagnosed with PDAC or pNET. Using specialized software, radiologists manually mapped regions of interest (ROIs) from computed tomography images and automatically extracted radiomics features. A total of 45 discriminative models were built by five selection algorithms and nine classification algorithms. The performances of the discriminative models were assessed by sensitivity, specificity and the area under receiver operating characteristic curve (AUC) in the training and validation datasets. Using the combination of Gradient Boosting Decision Tree (GBDT) as the selection algorithm and Random Forest (RF) as the classification algorithm, the optimal diagnostic ability with the highest AUC was presented in the training and validation datasets. The sensitivity, specificity and AUC of the model were 0.804, 0.973 and 0.971 in the training dataset and 0.742, 0.934 and 0.930 in the validation dataset, respectively. The combination of radiomics and multiple machine learning algorithms showed the potential ability to discriminate PDAC from pNET. We suggest that multi-algorithm modeling should be considered for similar studies in the future rather than using a single algorithm empirically.
Collapse
|
40
|
A histopathological study of artery wall involvement in pancreatic cancer surgery. Langenbecks Arch Surg 2022; 407:3501-3511. [DOI: 10.1007/s00423-022-02689-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/15/2022] [Indexed: 11/09/2022]
|
41
|
Wang S, Lin C, Kolomaya A, Ostdiek-Wille GP, Wong J, Cheng X, Lei Y, Liu C. Compute Tomography Radiomics Analysis on Whole Pancreas Between Healthy Individual and Pancreatic Ductal Adenocarcinoma Patients: Uncertainty Analysis and Predictive Modeling. Technol Cancer Res Treat 2022; 21:15330338221126869. [PMID: 36184987 PMCID: PMC9530578 DOI: 10.1177/15330338221126869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Radiomics is a rapidly growing field that quantitatively extracts image features
in a high-throughput manner from medical imaging. In this study, we analyzed the
radiomics features of the whole pancreas between healthy individuals and
pancreatic cancer patients, and we established a predictive model that can
distinguish cancer patients from healthy individuals based on these radiomics
features. Methods: We retrospectively collected venous-phase scans
of contrast-enhanced computed tomography (CT) images from 181 control subjects
and 85 cancer case subjects for radiomics analysis and predictive modeling. An
attending radiation oncologist delineated the pancreas for all the subjects in
the Varian Eclipse system, and we extracted 924 radiomics features using
PyRadiomics. We established a feature selection pipeline to exclude redundant or
unstable features. We randomly selected 189 cases (60 cancer and 129 control) as
the training set. The remaining 77 subjects (25 cancer and 52 control) as a test
set. We trained a Random Forest model utilizing the stable features to
distinguish the cancer patients from the healthy individuals on the training
dataset. We analyzed the performance of our best model by running 5-fold
cross-validations on the training dataset and applied our best model to the test
set. Results: We identified that 91 radiomics features are stable
against various uncertainty sources, including bin width, resampling, image
transformation, image noise, and segmentation uncertainty. Eight of the 91
features are nonredundant. Our final predictive model, using these 8 features,
has achieved a mean area under the receiver operating characteristic curve (AUC)
of 0.99 ± 0.01 on the training dataset (189 subjects) by cross-validation. The
model achieved an AUC of 0.910 on the independent test set (77 subjects) and an
accuracy of 0.935. Conclusion: CT-based radiomics analysis based on
the whole pancreas can distinguish cancer patients from healthy individuals, and
it could potentially become an early detection tool for pancreatic cancer.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Radiation Oncology, University of Nebraska Medical
Center, Omaha, NE, USA,Shuo Wang, PhD, Department of Radiation
Oncology, University of Nebraska Medical Center, 986861 Nebraska Medical Center,
Omaha, NE 68198, USA. Chi Lin, MD, PhD,
Department of Radiation Oncology, University of Nebraska Medical Center, 986861
Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chi Lin
- Department of Radiation Oncology, University of Nebraska Medical
Center, Omaha, NE, USA
| | - Alexander Kolomaya
- College of Medicine, University of Nebraska Medical
Center, Omaha, NE, USA
| | | | - Jeffrey Wong
- Department of Radiation Oncology, University of Nebraska Medical
Center, Omaha, NE, USA
| | - Xiaoyue Cheng
- Department of Mathematics, University of Nebraska
Omaha, Omaha, NE, USA
| | - Yu Lei
- Department of Radiation Oncology, Barrow Neurological
Institute, Phoenix, AZ, USA
| | | |
Collapse
|
42
|
Kitamura K, Esaki M, Sone M, Sugawara S, Hiraoka N, Nara S, Ban D, Takamoto T, Mizui T, Shimada K. Prognostic Impact of Radiological Splenic Artery Involvement in Pancreatic Ductal Adenocarcinoma of the Body and Tail. Ann Surg Oncol 2022; 29:7047-7058. [PMID: 35691957 DOI: 10.1245/s10434-022-11950-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/08/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Splenic artery (SpA) involvement heralds poor prognosis in pancreatic ductal adenocarcinoma (PDAC) of the body and tail but is not included in the resectability criteria. This study evaluated the prognostic impact of radiological SpA involvement in PDAC of the body and tail. METHODS Preoperative computed tomography images of patients who underwent distal pancreatectomy for resectable PDAC of the body and tail (n = 242) at our hospital between 2004 and 2018 were graded according to splenic vessel involvement status as clear, abutment, or encasement. Clinicopathological prognostic factors and overall survival (OS) and recurrence-free survival (RFS) rates were compared between the three groups. The prognostic value of radiological involvement status was assessed using Harrell's concordance statistic (C-index) and time-dependent receiver-operating characteristic curve analysis and compared with pathological findings. RESULTS The diagnostic concordance rate was 0.87 (weighted κ statistic). Prognosis worsened with progression from clear, abutment, to encasement status. SpA encasement (hazard ratio [HR] 1.97, p = 0.04) predicted poor OS in multivariate Cox hazard regression analysis. SpA abutment (HR 1.77, p = 0.017) and encasement (HR 1.86, p = 0.034) independently predicted poor RFS. Splenic vein abutment and encasement were not significant predictors of poor OS or RFS. SpA encasement without adjuvant chemotherapy had the poorest prognosis because of early distant metastasis. The prognostic value was higher for radiological SpA involvement than for pathological SpA invasion. CONCLUSIONS Radiological SpA involvement status is a meaningful and reproducible prognostic indicator that can be used preoperatively for determining the treatment strategy in PDAC of the body and tail.
Collapse
Affiliation(s)
- Kei Kitamura
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Minoru Esaki
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, Japan.
| | - Miyuki Sone
- Department of Diagnostic Radiology, National Cancer Center Hospital, Tokyo, Japan
| | - Shunsuke Sugawara
- Department of Diagnostic Radiology, National Cancer Center Hospital, Tokyo, Japan
| | - Nobuyoshi Hiraoka
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Satoshi Nara
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Daisuke Ban
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Takeshi Takamoto
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Takahiro Mizui
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Kazuaki Shimada
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
43
|
What Can We Learn About Pancreatic Adenocarcinoma from Imaging? Hematol Oncol Clin North Am 2022; 36:911-928. [DOI: 10.1016/j.hoc.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
44
|
Balzano V, Laurent E, Florence AM, Lecuyer AI, Lefebvre C, Heitzmann P, Hammel P, Lecomte T, Grammatico-Guillon L. Time interval from last visit to imaging diagnosis influences outcome in pancreatic adenocarcinoma: A regional population-based study on linked medico-administrative and clinical data. Ther Adv Med Oncol 2022; 14:17588359221113264. [PMID: 36090802 PMCID: PMC9449516 DOI: 10.1177/17588359221113264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Excessive waiting time intervals for the diagnosis and treatment of patients with pancreatic cancer can influence their prognosis but they remain unclear. The objective was to describe time intervals from the medical visit to diagnostic imaging and to treatment and their prognostic impact in pancreatic cancer in one French region. Methods: This retrospective observational multicentre study included all patients with pancreatic cancer seen for the first time in 2017 in multidisciplinary team meetings (MTMs), where clinical data were collected. A probabilistic matching with the medico-administrative data from the French national healthcare database (Système National des Données de Santé) was performed to define the care pathway from clinical presentation to the beginning of treatment. Median key time intervals were estimated for both resected and unresected tumours. Factors associated with 1-year survival were studied using Cox model. Results: A total of 324 patients (88% of total patients with MTM presentation) were matched and included: male 54%, mean age 72 years ±9.2, Eastern Cooperative Oncology Group (ECOG) PS > 1 19.5%, metastatic disease at diagnosis 47.4%, tumour resection 16%. At 1 year, 57% had died (65% in the unresected group and 17% in the resected group). The median time interval from the medical visit to diagnostic imaging was 15 days [Q1–Q3: 8–44]. After imaging, median time intervals to definite diagnosis and to first treatment were 11 and 20 days, respectively. Significant prognostic factors associated with the risk of death at 1 year were ECOG PS > 1 (hazard ratio (HR) 2.1 [1.4–3.0]), metastasis (HR 2.7 [1.9–3.9]), no tumour resection (HR 2.7 [1.3–5.6]) and time interval between the medical visit and diagnostic imaging ⩾25 days (HR 1.7 [1.2–2.3]). Conclusion: Delay in access to diagnostic imaging impacted survival in patients with pancreatic cancer, regardless of whether tumour resection had been performed.
Collapse
Affiliation(s)
- Vittoria Balzano
- OncoCentre, Cancer network of the Centre-Val de Loire region, Tours, France.,Gastroenterology and Digestive Oncology Department, Teaching Hospital of Tours, Tours, France
| | - Emeline Laurent
- Public Health Unit, Epidemiology (EpiDcliC), Teaching Hospital of Tours, Tours, France.,Research Unit EA7505 "Education, Ethics and Health", University of Tours, Tours, France
| | - Aline-Marie Florence
- Public Health Unit, Epidemiology (EpiDcliC), Teaching Hospital of Tours, Tours, France.,Department of Public Healht, Faculty of Medicine,University of Tours, France
| | - Anne-Isabelle Lecuyer
- Public Health Unit, Epidemiology (EpiDcliC), Teaching Hospital of Tours, Tours, France.,Research Unit EA7505 "Education, Ethics and Health", University of Tours, Tours, France
| | - Carole Lefebvre
- OncoCentre, Cancer network of the Centre-Val de Loire region, Tours, France
| | - Patrick Heitzmann
- OncoCentre, Cancer network of the Centre-Val de Loire region, Tours, France
| | - Pascal Hammel
- Digestive and Medical Oncology Department, Paul Brousse University Hospital, Villejuif, France.,Paris-Saclay University, Villejuif, France
| | - Thierry Lecomte
- OncoCentre, Cancer network of the Centre-Val de Loire region, Tours, France.,University of Tours, Faculty of Medicine, Tours, France.,Gastroenterology and Digestive Oncology Department, Teaching Hospital of Tours, Tours, France
| | - Leslie Grammatico-Guillon
- Department of Public Healht, Faculty of Medicine, University of Tours, France.,Public Health Unit, Epidemiology (EpiDcliC), Teaching Hospital of Tours, 2 Boulevard Tonnellé, 37044 Tours cedex 9, France
| |
Collapse
|
45
|
Shi S, Luo Y, Wang M, Lin Z, Song M, Li Z, Peng Z, Feng ST. Tumor fibrosis correlates with the survival of patients with pancreatic adenocarcinoma and is predictable using clinicoradiological features. Eur Radiol 2022; 32:6314-6326. [PMID: 35420301 DOI: 10.1007/s00330-022-08745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/06/2022] [Accepted: 03/14/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES To evaluate the prognostic value of fibrosis for patients with pancreatic adenocarcinoma (PDAC) and preoperatively predict fibrosis using clinicoradiological features. Tumor fibrosis plays an important role in the chemoresistance of PDAC. However, the prognostic value of tumor fibrosis remains contradiction and accurate prediction of tumor fibrosis is required. METHODS The study included 131 patients with PDAC who underwent first-line surgery. The prognostic value of fibrosis and rounded cutoff fibrosis points for median overall survival (OS) and disease-free survival (DFS) were determined using Cox regression and receiver operating characteristic (ROC) analyses. Then the whole cohort was randomly divided into training (n = 88) and validation (n = 43) sets. Binary logistic regression analysis was performed to select independent risk factors for fibrosis in the training set, and a nomogram was constructed. Nomogram performance was assessed using a calibration curve and decision curve analysis (DCA). RESULTS Hazard ratios of fibrosis for OS and DFS were 1.121 (95% confidence interval [CI]: 1.082-1.161) and 1.110 (95% CI: 1.067-1.155). ROC analysis identified 40% as the rounded cutoff fibrosis point for median OS and DFS. Tumor diameter, carbohydrate antigen 19-9 level, and peripancreatic tumor infiltration were independent risk factors; areas under the nomogram curve were 0.810 and 0.804 in the training and validation sets, respectively. The calibration curve indicated good agreement of the nomogram, and DCA demonstrated good clinical usefulness. CONCLUSIONS Tumor fibrosis was associated with poor OS and DFS in patients with PDAC. The nomogram incorporating clinicoradiological features was useful for preoperatively predicting tumor fibrosis. KEY POINTS • Tumor fibrosis is correlated with poor prognosis in patients with pancreatic adenocarcinoma. • Tumor fibrosis can be categorized according to its association with overall survival and disease-free survival. • A nomogram incorporating carbohydrate antigen 19-9 level, tumor diameter, and peripancreatic tumor infiltration is useful for preoperatively predicting tumor fibrosis.
Collapse
Affiliation(s)
- Siya Shi
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Second Zhongshan Road, Yuexiu District, Guangzhou, 510080, Guangdong, China
| | - Yanji Luo
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Second Zhongshan Road, Yuexiu District, Guangzhou, 510080, Guangdong, China
| | - Meng Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Second Zhongshan Road, Yuexiu District, Guangzhou, 510080, Guangdong, China
| | - Zhi Lin
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Second Zhongshan Road, Yuexiu District, Guangzhou, 510080, Guangdong, China
| | - Meiyi Song
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ziping Li
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Second Zhongshan Road, Yuexiu District, Guangzhou, 510080, Guangdong, China
| | - Zhenpeng Peng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Second Zhongshan Road, Yuexiu District, Guangzhou, 510080, Guangdong, China.
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Second Zhongshan Road, Yuexiu District, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
46
|
Marti-Bonmati L, Cerdá-Alberich L, Pérez-Girbés A, Díaz Beveridge R, Montalvá Orón E, Pérez Rojas J, Alberich-Bayarri A. Pancreatic cancer, radiomics and artificial intelligence. Br J Radiol 2022; 95:20220072. [PMID: 35687700 PMCID: PMC10996946 DOI: 10.1259/bjr.20220072] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/19/2022] [Accepted: 05/27/2022] [Indexed: 11/05/2022] Open
Abstract
Patients with pancreatic ductal adenocarcinoma (PDAC) are generally classified into four categories based on contrast-enhanced CT at diagnosis: resectable, borderline resectable, unresectable, and metastatic disease. In the initial grading and staging of PDAC, structured radiological templates are useful but limited, as there is a need to define the aggressiveness and microscopic disease stage of these tumours to ensure adequate treatment allocation. Quantitative imaging analysis allows radiomics and dynamic imaging features to provide information of clinical outcomes, and to construct clinical models based on radiomics signatures or imaging phenotypes. These quantitative features may be used as prognostic and predictive biomarkers in clinical decision-making, enabling personalised management of advanced PDAC. Deep learning and convolutional neural networks also provide high level bioinformatics tools that can help define features associated with a given aspect of PDAC biology and aggressiveness, paving the way to define outcomes based on these features. Thus, the prediction of tumour phenotype, treatment response and patient prognosis may be feasible by using such comprehensive and integrated radiomics models. Despite these promising results, quantitative imaging is not ready for clinical implementation in PDAC. Limitations include the instability of metrics and lack of external validation. Large properly annotated datasets, including relevant semantic features (demographics, blood markers, genomics), image harmonisation, robust radiomics analysis, clinically significant tasks as outputs, comparisons with gold-standards (such as TNM or pretreatment classifications) and fully independent validation cohorts, will be required for the development of trustworthy radiomics and artificial intelligence solutions to predict PDAC aggressiveness in a clinical setting.
Collapse
Affiliation(s)
- Luis Marti-Bonmati
- GIBI230 Research Group on Biomedical Imaging, Instituto de
Investigación Sanitaria La Fe,
Valencia, Spain
- Department of Radiology, Hospital Universitario y
Politécnico La Fe, Valencia,
Spain
| | - Leonor Cerdá-Alberich
- GIBI230 Research Group on Biomedical Imaging, Instituto de
Investigación Sanitaria La Fe,
Valencia, Spain
| | | | | | - Eva Montalvá Orón
- Department of Surgery, Hospital Universitario y
Politécnico La Fe, Valencia,
Spain
| | - Judith Pérez Rojas
- Department of Pathology, Hospital Universitario y
Politécnico La Fe, Valencia,
Spain
| | - Angel Alberich-Bayarri
- GIBI230 Research Group on Biomedical Imaging, Instituto de
Investigación Sanitaria La Fe,
Valencia, Spain
- Quantitative Imaging Biomarkers in Medicine, Quibim
SL, Valencia,
Spain
| |
Collapse
|
47
|
Nelson B, Barrord M, Wang K, Wages NA, Sudhoff M, Kharofa J. Relationship of dose to vascular target volumes and local failure in pancreatic cancer patients undergoing neoadjuvant chemoradiation. Front Oncol 2022; 12:906484. [PMID: 36119519 PMCID: PMC9470914 DOI: 10.3389/fonc.2022.906484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveThe objectives of this study were to evaluate whether dose to the vasculature is associated with local control after surgery in patients with borderline resectable (BLR) and resectable pancreatic cancer (PCA) receiving neoadjuvant radiation therapy (RT) and to identify a dose threshold for clinical use.MethodsPatients with BLR and resectable PCA treated with neoadjuvant RT were retrospectively reviewed. During this period, the institutional paradigm shifted from standard fractionation to hypofractionation/stereotactic body radiation therapy (SBRT). A vasculature clinical target volume (Vasc CTV) was contoured for each patient and defined as a 5-mm margin around the superior mesenteric artery (SMA) from its origin to the pancreatic head, the celiac artery from its origin to the level of the trifurcation and any involved vein. The Vasc CTV D95 was normalized to a 2-Gy equivalent dose to determine the optimal dose associated with optimal local failure-free survival (LFFS).ResultsForty-seven patients were included in the analysis. A Vasc CTV D95 of 32.7 Gy was the optimal cutoff for LFFS. Patients with Vasc CTV D95 Equivalent dose in 2 Gy per fraction (EQD2) >32.7 Gy had significantly longer LFFS compared to patients with Vasc CTV D95 EQD2 ≤32.7 Gy at 12 months (91% vs. 51%, respectively) and 24 months (86% vs. 12%, respectively). The median disease-free survival (DFS) for patients with EQD2 >32.7 Gy was 30.4 months compared to 14.0 months in patients with EQD2 ≤32.7 Gy (p = 0.01). There was no significant difference in overall survival (OS) between the two groups.ConclusionsDuring neoadjuvant treatment, dose to the Vasc CTV is associated with durability of local control (LC) after resection and should be intentionally included in the treatment volume with an EQD2 goal of 31–33 Gy.
Collapse
Affiliation(s)
- Bailey Nelson
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, United States
- *Correspondence: Bailey Nelson,
| | - Michelle Barrord
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, United States
| | - Kyle Wang
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, United States
| | - Nolan A. Wages
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Mickaela Sudhoff
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, United States
| | - Jordan Kharofa
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
48
|
Structured Reporting in Radiological Settings: Pitfalls and Perspectives. J Pers Med 2022; 12:jpm12081344. [PMID: 36013293 PMCID: PMC9409900 DOI: 10.3390/jpm12081344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/08/2022] [Accepted: 08/17/2022] [Indexed: 12/01/2022] Open
Abstract
Objective: The aim of this manuscript is to give an overview of structured reporting in radiological settings. Materials and Method: This article is a narrative review on structured reporting in radiological settings. Particularly, limitations and future perspectives are analyzed. RESULTS: The radiological report is a communication tool for the referring physician and the patients. It was conceived as a free text report (FTR) to allow radiologists to have their own individuality in the description of the radiological findings. However, this form could suffer from content, style, and presentation discrepancies, with a probability of transferring incorrect radiological data. Quality, datafication/quantification, and accessibility represent the three main goals in moving from FTRs to structured reports (SRs). In fact, the quality is related to standardization, which aims to improve communication and clarification. Moreover, a “structured” checklist, which allows all the fundamental items for a particular radiological study to be reported and permits the connection of the radiological data with clinical features, allowing a personalized medicine. With regard to accessibility, since radiological reports can be considered a source of research data, SR allows data mining to obtain new biomarkers and to help the development of new application domains, especially in the field of radiomics. Conclusions: Structured reporting could eliminate radiologist individuality, allowing a standardized approach.
Collapse
|
49
|
Moutinho-Ribeiro P, Batista IA, Quintas ST, Adem B, Silva M, Morais R, Peixoto A, Coelho R, Costa-Moreira P, Medas R, Lopes S, Vilas-Boas F, Baptista M, Dias-Silva D, Esteves AL, Martins F, Lopes J, Barroca H, Carneiro F, Macedo G, Melo SA. Exosomal glypican-1 is elevated in pancreatic cancer precursors and can signal genetic predisposition in the absence of endoscopic ultrasound abnormalities. World J Gastroenterol 2022; 28:4310-4327. [PMID: 36159010 PMCID: PMC9453765 DOI: 10.3748/wjg.v28.i31.4310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/30/2022] [Accepted: 06/26/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Individuals within specific risk groups for pancreatic ductal adenocarcinoma (PDAC) [mucinous cystic lesions (MCLs), hereditary risk (HR), and new-late onset diabetes mellitus (NLOD)] represent an opportunity for early cancer detection. Endoscopic ultrasound (EUS) is a premium image modality for PDAC screening and precursor lesion characterization. While no specific biomarker is currently clinically available for this purpose, glypican-1 (GPC1) is overexpressed in the circulating exosomes (crExos) of patients with PDAC compared with healthy subjects or those harboring benign pancreatic diseases.
AIM To evaluate the capacity of GPC1+ crExos to identify individuals at higher risk within these specific groups, all characterized by EUS.
METHODS This cross-sectional study with a prospective unicentric cohort included 88 subjects: 40 patients with MCL, 20 individuals with HR, and 20 patients with NLOD. A control group (CG) was submitted to EUS for other reasons than pancreatic pathology, with normal pancreas and absence of hereditary risk factors (n = 8). The inclusion period was between October 2016 and January 2019, and the study was approved by the Ethics Committee of Centro Hospitalar Universitário de São João, Porto, Portugal. All patients provided written informed consent. EUS and blood tests for quantification of GPC1+ crExos by flow cytometry and carbohydrate antigen 19-9 (CA 19-9) levels by ELISA were performed in all subjects. EUS-guided tissue acquisition was done whenever necessary. For statistical analysis, SPSS® 27.0 (IBM Corp., Armonk, NY, United States) version was used. All graphs were created using GraphPad Prism 7.00 (GraphPad Software, San Diego, CA, United States).
RESULTS Half of MCLs harbored worrisome features (WF) or high-risk stigmata (HRS). Pancreatic abnormalities were detected by EUS in 10.0% and 35.0% in HR and NLOD individuals, respectively, all considered non-malignant and “harmless.” Median levels of GPC1+ crExos were statistically different: MCL [99.4%, interquartile range (IQR): 94.9%-99.8%], HR (82.0%, IQR: 28.9%-98.2%), NLOD (12.6%, IQR: 5.2%-63.4%), and CG (16.2%, IQR: 6.6%-20.1%) (P < 0.0001). Median levels of CA 19-9 were within the normal range in all groups (standard clinical cut-off of 37 U/mL). Within HR, individuals with a positive history of cancer had higher median levels of GPC1+ crExos (97.9%; IQR: 61.7%-99.5%), compared to those without (59.7%; IQR: 26.3%-96.4%), despite no statistical significance (P = 0.21). Pancreatic cysts with WF/HRS were statistically associated with higher median levels of GPC1+ crExos (99.6%; IQR: 97.6%-99.8%) compared to those without (96.5%; IQR: 81.3%-99.5%) (P = 0.011), presenting an area under the receiver operating characteristic curve value of 0.723 (sensitivity 75.0% and specificity 67.7%, using a cut-off of 98.5%; P = 0.012).
CONCLUSION GPC1+ crExos may act as biomarker to support the diagnosis and stratification of PDAC precursor lesions, and in signaling individuals with genetic predisposition in the absence of EUS abnormalities.
Collapse
Affiliation(s)
- Pedro Moutinho-Ribeiro
- Serviço de Gastrenterologia, Centro Hospitalar Universitário de São João, Porto 4200, Portugal
- Faculty of Medicine, University of Porto, Porto 4200, Portugal
| | - Ines A Batista
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto 4200, Portugal
- IPATIMUP–Institute of Molecular Pathology and Immunology, University of Porto, Porto 4200, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto 4050, Portugal
| | - Sofia T Quintas
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto 4200, Portugal
- IPATIMUP–Institute of Molecular Pathology and Immunology, University of Porto, Porto 4200, Portugal
| | - Bárbara Adem
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto 4200, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto 4050, Portugal
| | - Marco Silva
- Serviço de Gastrenterologia, Centro Hospitalar Universitário de São João, Porto 4200, Portugal
- Faculty of Medicine, University of Porto, Porto 4200, Portugal
| | - Rui Morais
- Serviço de Gastrenterologia, Centro Hospitalar Universitário de São João, Porto 4200, Portugal
- Faculty of Medicine, University of Porto, Porto 4200, Portugal
| | - Armando Peixoto
- Serviço de Gastrenterologia, Centro Hospitalar Universitário de São João, Porto 4200, Portugal
- Faculty of Medicine, University of Porto, Porto 4200, Portugal
| | - Rosa Coelho
- Serviço de Gastrenterologia, Centro Hospitalar Universitário de São João, Porto 4200, Portugal
- Faculty of Medicine, University of Porto, Porto 4200, Portugal
| | - Pedro Costa-Moreira
- Serviço de Gastrenterologia, Centro Hospitalar Universitário de São João, Porto 4200, Portugal
- Faculty of Medicine, University of Porto, Porto 4200, Portugal
| | - Renato Medas
- Serviço de Gastrenterologia, Centro Hospitalar Universitário de São João, Porto 4200, Portugal
- Faculty of Medicine, University of Porto, Porto 4200, Portugal
| | - Susana Lopes
- Serviço de Gastrenterologia, Centro Hospitalar Universitário de São João, Porto 4200, Portugal
- Faculty of Medicine, University of Porto, Porto 4200, Portugal
| | - Filipe Vilas-Boas
- Serviço de Gastrenterologia, Centro Hospitalar Universitário de São João, Porto 4200, Portugal
- Faculty of Medicine, University of Porto, Porto 4200, Portugal
| | - Manuela Baptista
- Serviço de Cirurgia Geral, Centro Hospitalar Universitário de São João, Porto 4200, Portugal
| | - Diogo Dias-Silva
- Unidade de Saúde Familiar Serpa Pinto, ACeS Porto Ocidental, Porto 4250, Portugal
| | - Ana L Esteves
- Unidade de Saúde Familiar Serpa Pinto, ACeS Porto Ocidental, Porto 4250, Portugal
| | - Filipa Martins
- Unidade de Saúde Familiar Serpa Pinto, ACeS Porto Ocidental, Porto 4250, Portugal
| | - Joanne Lopes
- Serviço de Anatomia Patológica, Centro Hospitalar Universitário de São João, Porto 4200, Portugal
| | - Helena Barroca
- Serviço de Anatomia Patológica, Centro Hospitalar Universitário de São João, Porto 4200, Portugal
| | - Fátima Carneiro
- Faculty of Medicine, University of Porto, Porto 4200, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto 4200, Portugal
- IPATIMUP–Institute of Molecular Pathology and Immunology, University of Porto, Porto 4200, Portugal
- Serviço de Anatomia Patológica, Centro Hospitalar Universitário de São João, Porto 4200, Portugal
| | - Guilherme Macedo
- Serviço de Gastrenterologia, Centro Hospitalar Universitário de São João, Porto 4200, Portugal
- Faculty of Medicine, University of Porto, Porto 4200, Portugal
| | - Sonia A Melo
- Faculty of Medicine, University of Porto, Porto 4200, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto 4200, Portugal
- IPATIMUP–Institute of Molecular Pathology and Immunology, University of Porto, Porto 4200, Portugal
| |
Collapse
|
50
|
Wood LD, Canto MI, Jaffee EM, Simeone DM. Pancreatic Cancer: Pathogenesis, Screening, Diagnosis, and Treatment. Gastroenterology 2022; 163:386-402.e1. [PMID: 35398344 PMCID: PMC9516440 DOI: 10.1053/j.gastro.2022.03.056] [Citation(s) in RCA: 296] [Impact Index Per Article: 148.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/13/2022] [Accepted: 03/25/2022] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a clinically challenging cancer, due to both its late stage at diagnosis and its resistance to chemotherapy. However, recent advances in our understanding of the biology of PDAC have revealed new opportunities for early detection and targeted therapy of PDAC. In this review, we discuss the pathogenesis of PDAC, including molecular alterations in tumor cells, cellular alterations in the tumor microenvironment, and population-level risk factors. We review the current status of surveillance and early detection of PDAC, including populations at high risk and screening approaches. We outline the diagnostic approach to PDAC and highlight key treatment considerations, including how therapeutic approaches change with disease stage and targetable subtypes of PDAC. Recent years have seen significant improvements in our approaches to detect and treat PDAC, but large-scale, coordinated efforts will be needed to maximize the clinical impact for patients and improve overall survival.
Collapse
Affiliation(s)
- Laura D Wood
- Departments of Pathology and Oncology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Marcia Irene Canto
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth M Jaffee
- Sidney Kimmel Cancer Center, Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Diane M Simeone
- Departments of Surgery and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York
| |
Collapse
|