1
|
Mohamed G, Munir M, Rai A, Gaddam S. Pancreatic Cancer: Screening and Early Detection. Gastroenterol Clin North Am 2025; 54:205-221. [PMID: 39880528 DOI: 10.1016/j.gtc.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Pancreatic cancer, often diagnosed at advanced stages, has poor survival rates. Effective screening aims to detect the disease early, improving outcomes. Current guidelines recommend screening high-risk groups, including those with a family history or genetic predispositions, using methods like endoscopic ultrasound and MRI. The American Gastroenterological Association and other organizations advise annual surveillance for high-risk individuals, typically starting at the age of 50 or 10 years younger than the youngest affected relative. For certain genetic syndromes, such as Peutz-Jeghers syndrome or hereditary pancreatitis, screening may begin as early as the age of 35 to 40 years.
Collapse
Affiliation(s)
- Ghada Mohamed
- Department of Internal Medicine, Lahey Hospital & Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Malak Munir
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, ST, Suite 7705, Los Angeles, CA 90048, USA
| | - Amar Rai
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, ST, Suite 7705, Los Angeles, CA 90048, USA
| | - Srinivas Gaddam
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, ST, Suite 7705, Los Angeles, CA 90048, USA.
| |
Collapse
|
2
|
Puik JR, Poels TT, Hooijer GKJ, Cysouw MCF, Verheij J, Wilmink JW, Giovannetti E, Kazemier G, Sarasqueta AF, Oprea-Lager DE, Swijnenburg RJ. 18F-Prostate-Specific Membrane Antigen PET/CT imaging for potentially resectable pancreatic cancer (PANSCAN-2): a phase I/II study. Cancer Imaging 2025; 25:2. [PMID: 39810252 PMCID: PMC11734402 DOI: 10.1186/s40644-025-00822-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: 03/07/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Current diagnostic imaging modalities have limited ability to differentiate between malignant and benign pancreaticobiliary disease, and lack accuracy in detecting lymph node metastases. 18F-Prostate-Specific Membrane Antigen (PSMA) PET/CT is an imaging modality used for staging of prostate cancer, but has incidentally also identified PSMA-avid pancreatic lesions, histologically characterized as pancreatic ductal adenocarcinoma (PDAC). This phase I/II study aimed to assess the feasibility of 18F-PSMA PET/CT to detect PDAC. METHODS Seventeen patients with clinically resectable PDAC underwent 18F-PSMA PET/CT prior to surgical resection. Images were analyzed both visually and (semi)quantitatively by deriving the maximum standardized uptake value (SUVmax) and tumor-to-background ratio (TBR). TBR was defined as the ratio between SUVmax of the primary tumor divided by SUVmax of the aortic blood pool. Finally, tracer uptake on PET was correlated to tissue expression of PSMA in surgical specimens. RESULTS Out of 17 PSMA PET/CT scans, 13 scans demonstrated positive PSMA tracer uptake, with a mean SUVmax of 5.0 ± 1.3. The suspected primary tumor was detectable (TBR ≥ 2) with a mean TBR of 3.3 ± 1.3. For histologically confirmed PDAC, mean SUVmax and mean TBR were 4.9 ± 1.2 and 3.3 ± 1.5, respectively. Although eight patients had histologically confirmed regional lymph node metastases and two patients had distant metastases, none of these metastases demonstrated 18F-PSMA uptake. There was no correlation between 18F-PSMA PET/CT SUVmax and tissue expression of PSMA in surgical specimens. CONCLUSIONS 18F-PSMA PET/CT was able to detect several pancreaticobiliary cancers, including PDAC. However, uptake was generally low, not specific to PDAC and no tracer uptake was observed in lymph node or distant metastases. The added value of PSMA PET in this setting appears to be limited. TRIAL REGISTRATION The trial is registered as PANSCAN-2 in the European Clinical Trials Database (EudraCT number: 2020-002185-14).
Collapse
Affiliation(s)
- Jisce R Puik
- Department of Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Thomas T Poels
- Department of Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Gerrit K J Hooijer
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Pathology, UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Matthijs C F Cysouw
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Joanne Verheij
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Pathology, UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Johanna W Wilmink
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Elisa Giovannetti
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, Pisa, Italy
| | - Geert Kazemier
- Department of Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Arantza Farina Sarasqueta
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Pathology, UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Daniela E Oprea-Lager
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Rutger-Jan Swijnenburg
- Department of Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands.
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
- Department of Surgery, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Javed S, Qureshi TA, Wang L, Azab L, Gaddam S, Pandol SJ, Li D. An insight to PDAC tumor heterogeneity across pancreatic subregions using computed tomography images. Front Oncol 2024; 14:1378691. [PMID: 39600638 PMCID: PMC11588633 DOI: 10.3389/fonc.2024.1378691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is an exceptionally deadly form of pancreatic cancer with an extremely low survival rate. From diagnosis to treatment, PDAC is highly challenging to manage. Studies have demonstrated that PDAC tumors in distinct regions of the pancreas exhibit unique characteristics, influencing symptoms, treatment responses, and survival rates. Gaining insight into the heterogeneity of PDAC tumors based on their location in the pancreas can significantly enhance overall management of PDAC. Previous studies have explored PDAC tumor heterogeneity across pancreatic subregions based on their genetic and molecular profiles through biopsy-based histologic assessment. However, biopsy examinations are highly invasive and impractical for large populations. Abdominal imaging, such as Computed Tomography (CT) offers a completely non-invasive means to evaluate PDAC tumor heterogeneity across pancreatic subregions and an opportunity to correlate image feature of tumors with treatment outcome and monitoring. In this study, we explored the inter-tumor heterogeneity in PDAC tumors across three primary pancreatic subregions: the head, body, and tail. Utilizing contrast-enhanced abdominal CT scans and a thorough radiomic analysis of PDAC tumors, several morphological and textural tumor features were identified to be notably different between tumors in the head and those in the body and tail regions. To validate the significance of the identified features, a machine learning ML model was trained to automatically classify PDAC tumors into their respective regions i.e. head or body/tail subregion using their CT features. The study involved 200 CT abdominal scans, with 100 used for radiomic analysis and model training, and the remaining 100 for model testing. The ML model achieved an average classification accuracy, sensitivity, and specificity of 87%, 86%, and 88% on the testing scans respectively. Evaluating the heterogeneity of PDAC tumors across pancreatic subregions provides valuable insights into tumor composition and has the potential to enhance diagnosis and personalize treatment based on tumor characteristics and location.
Collapse
Affiliation(s)
- Sehrish Javed
- Cedars Sinai Medical Center, Los Angeles, CA, United States
| | | | | | | | | | | | - Debiao Li
- Cedars Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
4
|
Kotb A, Hafeji Z, Jesry F, Lintern N, Pathak S, Smith AM, Lutchman KRD, de Bruin DM, Hurks R, Heger M, Khaled YS. Intra-Operative Tumour Detection and Staging in Pancreatic Cancer Surgery: An Integrative Review of Current Standards and Future Directions. Cancers (Basel) 2024; 16:3803. [PMID: 39594758 PMCID: PMC11592681 DOI: 10.3390/cancers16223803] [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: 06/09/2024] [Revised: 10/15/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Surgical resection for pancreatic ductal adenocarcinoma (PDAC) entails the excision of the primary tumour and regional lymphadenectomy. This traditional strategy is challenged by the high rate of early recurrence, suggesting inadequate disease staging. Novel methods of intra-operative staging are needed to allow surgical resection to be tailored to the disease's biology. METHODS A search of published articles on the PubMed and Embase databases was performed using the terms 'pancreas' OR 'pancreatic' AND 'intra-operative staging/detection' OR 'guided surgery'. Articles published between January 2000 and June 2023 were included. Technologies that offered intra-operative staging and tailored treatment were curated and summarised in the following integrative review. RESULTS lymph node (LN) mapping and radioimmunoguided surgery have shown promising results but lacked practicality to facilitate real-time intra-operative staging for PDAC. Fluorescence-guided surgery (FGS) offers high contrast and sensitivity, enabling the identification of cancerous tissue and positive LNs with improved precision following intravenous administration of a fluorescent agent. The unique properties of optical coherence tomography and ultrasound elastography lend themselves to be platforms for virtual biopsy intra-operatively. CONCLUSIONS Accurate intra-operative staging of PDAC, localisation of metastatic LNs, and identification of extra-pancreatic disease remain clinically unmet needs under current detection methods and staging standards. Tumour-specific FGS combined with other diagnostic and therapeutic modalities could improve tumour detection and staging in patients with PDAC.
Collapse
Affiliation(s)
- Ahmed Kotb
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| | - Zaynab Hafeji
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| | - Fadel Jesry
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| | - Nicole Lintern
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| | - Samir Pathak
- The Pancreato-Biliary Unit, St James’s University Teaching Hospital, Leeds LS9 7TF, UK
| | - Andrew M. Smith
- The Pancreato-Biliary Unit, St James’s University Teaching Hospital, Leeds LS9 7TF, UK
| | - Kishan R. D. Lutchman
- Department of Surgery, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands
- Department of Biomedical Engineering and Physics, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands
| | - Daniel M. de Bruin
- Department of Biomedical Engineering and Physics, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands
| | - Rob Hurks
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China
| | - Yazan S. Khaled
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
- The Pancreato-Biliary Unit, St James’s University Teaching Hospital, Leeds LS9 7TF, UK
| |
Collapse
|
5
|
Brandl A, Lundon D, Siriwardena AK, Sochorova D, Ceelen W, Besselink M, Soreide K, Stättner S. Surgical management of pancreatic neuroendocrine tumors - An EYSAC and E-AHPBA international survey of current practice. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108544. [PMID: 39059195 DOI: 10.1016/j.ejso.2024.108544] [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: 05/07/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024]
Abstract
INTRODUCTION Pancreatic neuroendocrine tumors (pNET) exhibit a wide spectrum of clinical behavior, which makes their assessment and management quite challenging. The purpose of this study was to comprehensively assess the existing treatment landscape for patients with pNET. MATERIALS AND METHODS The study was conducted with the support of the ESSO-EYSAC Research Academy in collaboration with the E-AHPBA. An online survey was distributed via email and social media to surgical networks across Europe and beyond (September 1-30, 2023). RESULTS Overall, 155 complete responses were obtained. A specialized NET tumor board was present at the institutions of 94 (61 %) of the study participants. The most frequently applied guidelines were from ENETS (n = 97; 63 %), NCCN (n = 74; 48 %), and ESMO (n = 53; 34 %). For resectability, similar criteria as in pancreatic ductal adenocarcinoma were used by 111 (72 %) participants, even though 116 (75 %) participants believed that pNET/pNEC should have their own resectability criteria. Most respondents used somatostatin analogues (n = 126; 81 %) and chemotherapy (n = 85; 55 %) as neoadjuvant treatments, followed by molecularly targeted agents (n = 45; 29 %) and PRRT (n = 37; 24 %). Only 17 (11 %) participants agreed/strongly agreed that the management of pNET/pNEC is sufficiently addressed in surgical education programs. CONCLUSION This international survey highlighted areas for improvement in the care of pNET, namely the lack of pNET-specific resectability criteria and educational programs addressing pNET management.
Collapse
Affiliation(s)
- Andreas Brandl
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Germany.
| | - Dara Lundon
- Department of Urology, Icahn School of Medicine at Mount Sinai Hospitals, New York, United States
| | - Ajith K Siriwardena
- Regional Hepato-Pancreato-Biliary Unit, Manchester Royal Infirmary, Manchester, UK
| | - Dana Sochorova
- Department of Surgery, Tomas Bata Regional Hospital, Zlin, Czech Republic
| | - Wim Ceelen
- Department of GI Surgery, Ghent University Hospital, and Cancer Research Institute Ghent (CRIG), Belgium
| | - Marc Besselink
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands; Cancer Center Amsterdam, the Netherlands
| | - Kjetil Soreide
- Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden; Department of Gastrointestinal Surgery, HPB Unit, Stavanger University Hospital, Stavanger, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Stefan Stättner
- Department of General, Visceral and Vascular Surgery, Salzkammergutklinikum, Vöcklabruck, Austria
| |
Collapse
|
6
|
Chen Y, Ma C, Yang P, Mao K, Gao Y, Chen L, Wang Z, Bian Y, Shao C, Lu J. Values of apparent diffusion coefficient in pancreatic cancer patients receiving neoadjuvant therapy. BMC Cancer 2024; 24:1160. [PMID: 39294623 PMCID: PMC11412028 DOI: 10.1186/s12885-024-12934-y] [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: 01/24/2024] [Accepted: 09/11/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND To investigate the values of apparent diffusion coefficient (ADC) for the treatment response evaluation in pancreatic cancer (PC) patients receiving neoadjuvant therapy (NAT). METHODS This study included 103 NAT patients with histologically proven PC. ADC maps were generated using monoexponential diffusion-weighted imaging (b values: 50, 800 s/mm2). Tumors' minimum, maximum, and mean ADCs were measured and compared pre- and post-NAT. Variations in ADC values measured between pre- and post-NAT completion for NAT methods (chemotherapy, chemoradiotherapy), tumor locations (head/neck, body/tail), tumor regression grade (TRG) levels (0-2, 3), N stages (N0, N1/N2) and tumor resection margin status (R0, R1), were further analyzed. RESULTS The minimum, maximum, and mean ADC values all increased dramatically after NAT, rising from 23.4 to 25.4% (all p < 0.001): mean (average: 1.626 × 10- 3 mm2/s vs. 1.315 × 10- 3 mm2/s), minimum (median: 1.274 × 10- 3 mm2/s vs. 1.034 × 10- 3 mm2/s), and maximum (average: 1.981 × 10- 3 mm2/s vs. 1.580 × 10- 3 mm2/s). The ADCs between the subgroups of all the criteria under investigation did not differ significantly for the minimum, maximum, or mean values pre- or post-NAT (P = 0.08 to 1.00). In the patients with borderline resectable PC (n = 47), the rate of tumor size changes after NAT was correlated with the pre-NAT mean ADC values (Spearman's coefficient: 0.288, P = 0.049). CONCLUSIONS The ADC values of PC increased significantly following NAT; however, the percentage increases failed to provide any predictive value for the resection margin status or TRG levels.
Collapse
Affiliation(s)
- Yufei Chen
- College of Electronic and Information Engineering, Tongji University, Shanghai, China
| | - Chao Ma
- College of Electronic and Information Engineering, Tongji University, Shanghai, China.
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China.
| | - Panpan Yang
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Kuanzheng Mao
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yisha Gao
- Department of Pathology, Changhai Hospital of Shanghai, Naval Medical University, Shanghai, China
| | - Luguang Chen
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Zhen Wang
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Yun Bian
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Chengwei Shao
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Jianping Lu
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| |
Collapse
|
7
|
Bugazia D, Al-Najjar E, Esmail A, Abdelrahim S, Abboud K, Abdelrahim A, Umoru G, Rayyan HA, Abudayyeh A, Al Moustafa AE, Abdelrahim M. Pancreatic ductal adenocarcinoma: the latest on diagnosis, molecular profiling, and systemic treatments. Front Oncol 2024; 14:1386699. [PMID: 39011469 PMCID: PMC11247645 DOI: 10.3389/fonc.2024.1386699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/30/2024] [Indexed: 07/17/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is currently the fourth leading cause of death in the United States and is expected to be ranked second in the next 10 years due to poor prognosis and a rising incidence. Distant metastatic PDAC is associated with the worst prognosis among the different phases of PDAC. The diagnostic options for PDAC are convenient and available for staging, tumor response evaluation, and management of resectable or borderline resectable PDAC. However, imaging is crucial in PDAC diagnosis, monitoring, resectability appraisal, and response evaluation. The advancement of medical technologies is evolving, hence the use of imaging in PDAC treatment options has grown as well as the utilization of ctDNA as a tumor marker. Treatment options for metastatic PDAC are minimal with the primary goal of therapy limited to symptom relief or palliation, especially in patients with low functional capacity at the point of diagnosis. Molecular profiling has shown promising potential solutions that would push the treatment boundaries for patients with PDAC. In this review, we will discuss the latest updates from evidence-based guidelines regarding diagnosis, therapy response evaluation, prognosis, and surveillance, as well as illustrating novel therapies that have been recently investigated for PDAC, in addition to discussing the molecular profiling advances in PDAC.
Collapse
Affiliation(s)
- Doaa Bugazia
- Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Ebtesam Al-Najjar
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX, United States
| | - Abdullah Esmail
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX, United States
| | - Saifudeen Abdelrahim
- Challenge Early College HS, Houston Community College, Houston, TX, United States
| | - Karen Abboud
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX, United States
| | | | - Godsfavour Umoru
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX, United States
| | - Hashem A Rayyan
- Department of Medicine, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Ala Abudayyeh
- Section of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Maen Abdelrahim
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
8
|
Ramaekers M, Viviers CGA, Hellström TAE, Ewals LJS, Tasios N, Jacobs I, Nederend J, Sommen FVD, Luyer MDP. Improved Pancreatic Cancer Detection and Localization on CT Scans: A Computer-Aided Detection Model Utilizing Secondary Features. Cancers (Basel) 2024; 16:2403. [PMID: 39001465 PMCID: PMC11240790 DOI: 10.3390/cancers16132403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
The early detection of pancreatic ductal adenocarcinoma (PDAC) is essential for optimal treatment of pancreatic cancer patients. We propose a tumor detection framework to improve the detection of pancreatic head tumors on CT scans. In this retrospective research study, CT images of 99 patients with pancreatic head cancer and 98 control cases from the Catharina Hospital Eindhoven were collected. A multi-stage 3D U-Net-based approach was used for PDAC detection including clinically significant secondary features such as pancreatic duct and common bile duct dilation. The developed algorithm was evaluated using a local test set comprising 59 CT scans. The model was externally validated in 28 pancreatic cancer cases of a publicly available medical decathlon dataset. The tumor detection framework achieved a sensitivity of 0.97 and a specificity of 1.00, with an area under the receiver operating curve (AUROC) of 0.99, in detecting pancreatic head cancer in the local test set. In the external test set, we obtained similar results, with a sensitivity of 1.00. The model provided the tumor location with acceptable accuracy obtaining a DICE Similarity Coefficient (DSC) of 0.37. This study shows that a tumor detection framework utilizing CT scans and secondary signs of pancreatic cancer can detect pancreatic tumors with high accuracy.
Collapse
Affiliation(s)
- Mark Ramaekers
- Department of Surgery, Catharina Cancer Institute, Catharina Hospital Eindhoven, EJ 5623 Eindhoven, The Netherlands
| | - Christiaan G A Viviers
- Department of Electrical Engineering, Eindhoven University of Technology, AZ 5612 Eindhoven, The Netherlands
| | - Terese A E Hellström
- Department of Electrical Engineering, Eindhoven University of Technology, AZ 5612 Eindhoven, The Netherlands
| | - Lotte J S Ewals
- Department of Radiology, Catharina Cancer Institute, Catharina Hospital Eindhoven, EJ 5623 Eindhoven, The Netherlands
| | - Nick Tasios
- Department of Hospital Services and Informatics, Philips Research, AE 5656 Eindhoven, The Netherlands
| | - Igor Jacobs
- Department of Hospital Services and Informatics, Philips Research, AE 5656 Eindhoven, The Netherlands
| | - Joost Nederend
- Department of Radiology, Catharina Cancer Institute, Catharina Hospital Eindhoven, EJ 5623 Eindhoven, The Netherlands
| | - Fons van der Sommen
- Department of Electrical Engineering, Eindhoven University of Technology, AZ 5612 Eindhoven, The Netherlands
| | - Misha D P Luyer
- Department of Surgery, Catharina Cancer Institute, Catharina Hospital Eindhoven, EJ 5623 Eindhoven, The Netherlands
| |
Collapse
|
9
|
Egorov V, Kim P, Dzigasov S, Kondratiev E, Sorokin A, Kolygin A, Vyborniy M, Bolshakov G, Popov P, Demchenkova A, Dakhtler T. Pancreatectomy with En Bloc Superior Mesenteric Vein and All Its Tributaries Resection without PV/SMV Reconstruction for "Low" Locally Advanced Pancreatic Head Cancer. Cancers (Basel) 2024; 16:2234. [PMID: 38927939 PMCID: PMC11202096 DOI: 10.3390/cancers16122234] [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: 05/14/2024] [Revised: 06/09/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
The "vein definition" for locally advanced pancreatic ductal adenocarcinoma (LA PDAC) assumes portal-to-superior mesenteric vein (PV/SMV) unreconstructability due to tumor involvement or occlusion. Radical pancreatectomies with SMV resection without PV/SMV reconstruction are scarcely discussed in the literature. Retrospective analysis of 19 radical pancreatectomies for "low" LA PDAC with SMV and all its tributaries resection without PV/SMV reconstruction has shown zero mortality; overall morbidity-56%; Dindo-Clavien-3-10.5%; R0-rate-82%; mean operative procedure time-355 ± 154 min; mean blood loss-330 ± 170 mL; delayed gastric emptying-25%; and clinically relevant postoperative pancreatic fistula-8%. In three cases, surgery was associated with superior mesenteric (n2) and common hepatic artery (n1) resection. Surgery was completed without vein reconstruction (n13) and with inferior mesenteric-to-splenic anastomosis (n6). There were no cases of liver, gastric, or intestinal ischemia. A specific complication of the SMV resection without reconstruction was 2-3 days-long intestinal edema (48%). Median overall survival was 25 months, and median progression-free survival was 18 months. All the relapses, except two, were distant. The possibility of successful SMV resection without PV/SMV reconstruction can be predicted before surgery by CT-based reconstructions. The mandatory anatomical conditions for the procedure were as follows: (1) preserved SMV-SV confluence; (2) occluded SMV for any reason (tumor or thrombus); (3) well-developed inferior mesenteric vein collaterals with dilated intestinal veins; (4) no right-sided vein collaterals; and (5) no varices in the upper abdomen. Conclusion: "Low" LA PDACs involving SMV with all its tributaries can be radically and safely resected in highly and specifically selected cases without PV/SMV reconstruction with an acceptable survival rate.
Collapse
Affiliation(s)
- Viacheslav Egorov
- Ilyinskaya Hospital, 143421 Moscow, Russia; (P.K.); (S.D.); (E.K.); (A.K.); (M.V.); (G.B.); (P.P.); (A.D.); (T.D.)
- Burnasyan State Research Center of the Federal Medical Biological Agency, 119435 Moscow, Russia
| | - Pavel Kim
- Ilyinskaya Hospital, 143421 Moscow, Russia; (P.K.); (S.D.); (E.K.); (A.K.); (M.V.); (G.B.); (P.P.); (A.D.); (T.D.)
| | - Soslan Dzigasov
- Ilyinskaya Hospital, 143421 Moscow, Russia; (P.K.); (S.D.); (E.K.); (A.K.); (M.V.); (G.B.); (P.P.); (A.D.); (T.D.)
| | - Eugeny Kondratiev
- Ilyinskaya Hospital, 143421 Moscow, Russia; (P.K.); (S.D.); (E.K.); (A.K.); (M.V.); (G.B.); (P.P.); (A.D.); (T.D.)
- Radiology Department, Vishnevsky National Medical Research Center of Surgery, 117997 Moscow, Russia
| | - Alexander Sorokin
- Department of Mathematical Methods in Economics, Plekhanov Russian University of Economics, 117997 Moscow, Russia;
| | - Alexey Kolygin
- Ilyinskaya Hospital, 143421 Moscow, Russia; (P.K.); (S.D.); (E.K.); (A.K.); (M.V.); (G.B.); (P.P.); (A.D.); (T.D.)
| | - Mikhail Vyborniy
- Ilyinskaya Hospital, 143421 Moscow, Russia; (P.K.); (S.D.); (E.K.); (A.K.); (M.V.); (G.B.); (P.P.); (A.D.); (T.D.)
| | - Grigoriy Bolshakov
- Ilyinskaya Hospital, 143421 Moscow, Russia; (P.K.); (S.D.); (E.K.); (A.K.); (M.V.); (G.B.); (P.P.); (A.D.); (T.D.)
| | - Pavel Popov
- Ilyinskaya Hospital, 143421 Moscow, Russia; (P.K.); (S.D.); (E.K.); (A.K.); (M.V.); (G.B.); (P.P.); (A.D.); (T.D.)
| | - Anna Demchenkova
- Ilyinskaya Hospital, 143421 Moscow, Russia; (P.K.); (S.D.); (E.K.); (A.K.); (M.V.); (G.B.); (P.P.); (A.D.); (T.D.)
| | - Tatiana Dakhtler
- Ilyinskaya Hospital, 143421 Moscow, Russia; (P.K.); (S.D.); (E.K.); (A.K.); (M.V.); (G.B.); (P.P.); (A.D.); (T.D.)
| |
Collapse
|
10
|
Pande R, Liu W, Raza SS, Papamichail M, Suthananthan AE, Bartlett DC, Marudanayagam R, Dasari BVM, Sutcliffe RP, Roberts KJ, Wadhwani S, Chatzizacharias N. Staging Computed Tomography Parameters Predict the Need for Vein Resection during Pancreaticoduodenectomy in Resectable Pancreatic Ductal Adenocarcinoma. Diagnostics (Basel) 2024; 14:135. [PMID: 38248012 PMCID: PMC10814156 DOI: 10.3390/diagnostics14020135] [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: 12/11/2023] [Revised: 12/30/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Surgery-first approach is the current standard of care for resectable pancreatic ductal adenocarcinoma (PDAC), and a proportion of these cases will require venous resection. This study aimed to identify parameters on staging computed tomography (CT) that predict the need for venous resection during pancreaticoduodenectomy (PD) for resectable PDAC. METHODS We conducted a retrospective analysis of prospectively collected data on patients who underwent PD for resectable staged PDAC (as per NCCN criteria) between 2011 and 2020. Staging CTs were independently reviewed by two specialist radiologists blinded to the clinical outcomes. Univariate and multivariate risk analyses were performed. RESULTS In total, 296 PDs were included. Venous resection was performed in 62 (21%) cases. There was a higher rate of resection margin positivity in the vein resection group (72.6% vs. 48.7%, p = 0.001). Tumour at the neck of the pancreas, superior mesenteric vein involvement of ≥10 mm and pancreatic duct dilatation were identified as independent predictors for venous resection. DISCUSSION Staging CT parameters can predict the need for venous resection during PD for resectable cases of PDAC. This may assist in surgical planning, patient selection and counselling. Future efforts should concentrate on validating these results or identifying additional predictors in a multicentre and prospective setting.
Collapse
Affiliation(s)
- Rupaly Pande
- Department of HPB and Liver Transplant Surgery, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK; (R.P.); (S.S.R.); (M.P.); (A.E.S.); (D.C.B.); (R.M.); (B.V.M.D.); (R.P.S.); (K.J.R.)
| | - Wingyan Liu
- Department of Radiology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK; (W.L.); (S.W.)
| | - Syed S. Raza
- Department of HPB and Liver Transplant Surgery, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK; (R.P.); (S.S.R.); (M.P.); (A.E.S.); (D.C.B.); (R.M.); (B.V.M.D.); (R.P.S.); (K.J.R.)
| | - Michail Papamichail
- Department of HPB and Liver Transplant Surgery, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK; (R.P.); (S.S.R.); (M.P.); (A.E.S.); (D.C.B.); (R.M.); (B.V.M.D.); (R.P.S.); (K.J.R.)
| | - Arul E. Suthananthan
- Department of HPB and Liver Transplant Surgery, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK; (R.P.); (S.S.R.); (M.P.); (A.E.S.); (D.C.B.); (R.M.); (B.V.M.D.); (R.P.S.); (K.J.R.)
| | - David C. Bartlett
- Department of HPB and Liver Transplant Surgery, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK; (R.P.); (S.S.R.); (M.P.); (A.E.S.); (D.C.B.); (R.M.); (B.V.M.D.); (R.P.S.); (K.J.R.)
| | - Ravi Marudanayagam
- Department of HPB and Liver Transplant Surgery, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK; (R.P.); (S.S.R.); (M.P.); (A.E.S.); (D.C.B.); (R.M.); (B.V.M.D.); (R.P.S.); (K.J.R.)
| | - Bobby V. M. Dasari
- Department of HPB and Liver Transplant Surgery, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK; (R.P.); (S.S.R.); (M.P.); (A.E.S.); (D.C.B.); (R.M.); (B.V.M.D.); (R.P.S.); (K.J.R.)
| | - Robert P. Sutcliffe
- Department of HPB and Liver Transplant Surgery, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK; (R.P.); (S.S.R.); (M.P.); (A.E.S.); (D.C.B.); (R.M.); (B.V.M.D.); (R.P.S.); (K.J.R.)
| | - Keith J. Roberts
- Department of HPB and Liver Transplant Surgery, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK; (R.P.); (S.S.R.); (M.P.); (A.E.S.); (D.C.B.); (R.M.); (B.V.M.D.); (R.P.S.); (K.J.R.)
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Sharan Wadhwani
- Department of Radiology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK; (W.L.); (S.W.)
| | - Nikolaos Chatzizacharias
- Department of HPB and Liver Transplant Surgery, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK; (R.P.); (S.S.R.); (M.P.); (A.E.S.); (D.C.B.); (R.M.); (B.V.M.D.); (R.P.S.); (K.J.R.)
- College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
11
|
Del Chiaro M, Sugawara T, Karam SD, Messersmith WA. Advances in the management of pancreatic cancer. BMJ 2023; 383:e073995. [PMID: 38164628 DOI: 10.1136/bmj-2022-073995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Pancreatic cancer remains among the malignancies with the worst outcomes. Survival has been improving, but at a slower rate than other cancers. Multimodal treatment, including chemotherapy, surgical resection, and radiotherapy, has been under investigation for many years. Because of the anatomical characteristics of the pancreas, more emphasis on treatment selection has been placed on local extension into major vessels. Recently, the development of more effective treatment regimens has opened up new treatment strategies, but urgent research questions have also become apparent. This review outlines the current management of pancreatic cancer, and the recent advances in its treatment. The review discusses future treatment pathways aimed at integrating novel findings of translational and clinical research.
Collapse
Affiliation(s)
- Marco Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, USA
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Toshitaka Sugawara
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sana D Karam
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Wells A Messersmith
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
12
|
Kos-Kudła B, Castaño JP, Denecke T, Grande E, Kjaer A, Koumarianou A, de Mestier L, Partelli S, Perren A, Stättner S, Valle JW, Fazio N. European Neuroendocrine Tumour Society (ENETS) 2023 guidance paper for nonfunctioning pancreatic neuroendocrine tumours. J Neuroendocrinol 2023; 35:e13343. [PMID: 37877341 DOI: 10.1111/jne.13343] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023]
Abstract
This ENETS guidance paper for well-differentiated nonfunctioning pancreatic neuroendocrine tumours (NF-Pan-NET) has been developed by a multidisciplinary working group, and provides up-to-date and practical advice on the management of these tumours. Using the extensive experience of centres treating patients with NF-Pan-NEN, the authors of this guidance paper discuss 10 troublesome questions in everyday clinical practice. Our many years of experience in this field are still being verified in the light of the results of new clinical, which set new ways of proceeding in NEN. The treatment of NF-Pan-NEN still requires a decision of a multidisciplinary team of specialists in the field of neuroendocrine neoplasms.
Collapse
Affiliation(s)
- Beata Kos-Kudła
- Department of Endocrinology and Neuroendocrine Tumours, Department of Pathophysiology and Endocrinology, Medical University of Silesia, Katowice, Poland
| | - Justo P Castaño
- Maimonides Biomedical Research Institute of Córdoba, University of Córdoba, Hospital Universitario Reina Sofía, Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Córdoba, Spain
| | - Timm Denecke
- Department of Diagnostic and Interventional Radiology, University Medical Centre Leipzig, Leipzig, Germany
| | - Enrique Grande
- Medical Oncology Department, MD Anderson Cancer Centre Madrid, Madrid, Spain
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine and Cluster for Molecular Imaging, Copenhagen University Hospital - Righospitalet and Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Louis de Mestier
- Université Paris-Cité, Department of Pancreatology and Digestive Oncology, Beaujon Hospital (APHP.Nord) and INSERM U1149, Paris, France
| | - Stefano Partelli
- Pancreatic Translational and Clinical Research Centre, Pancreatic and Transplant Surgery Unit, Vita-Salute San Raffaele University, Milan, Italy
| | - Aurel Perren
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Stefan Stättner
- Department of General, Visceral and Vascular Surgery, Salzkammergut Klinikum, OÖG, Vöcklabruck, Austria
| | - Juan W Valle
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumours, European Institute of Oncology (IEO), IRCCS, Milan, Italy
| |
Collapse
|
13
|
Wu Y, Seufert I, Al-Shaheri FN, Kurilov R, Bauer AS, Manoochehri M, Moskalev EA, Brors B, Tjaden C, Giese NA, Hackert T, Büchler MW, Hoheisel JD. DNA-methylation signature accurately differentiates pancreatic cancer from chronic pancreatitis in tissue and plasma. Gut 2023; 72:2344-2353. [PMID: 37709492 PMCID: PMC10715533 DOI: 10.1136/gutjnl-2023-330155] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/31/2023] [Indexed: 09/16/2023]
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy. Differentiation from chronic pancreatitis (CP) is currently inaccurate in about one-third of cases. Misdiagnoses in both directions, however, have severe consequences for patients. We set out to identify molecular markers for a clear distinction between PDAC and CP. DESIGN Genome-wide variations of DNA-methylation, messenger RNA and microRNA level as well as combinations thereof were analysed in 345 tissue samples for marker identification. To improve diagnostic performance, we established a random-forest machine-learning approach. Results were validated on another 48 samples and further corroborated in 16 liquid biopsy samples. RESULTS Machine-learning succeeded in defining markers to differentiate between patients with PDAC and CP, while low-dimensional embedding and cluster analysis failed to do so. DNA-methylation yielded the best diagnostic accuracy by far, dwarfing the importance of transcript levels. Identified changes were confirmed with data taken from public repositories and validated in independent sample sets. A signature of six DNA-methylation sites in a CpG-island of the protein kinase C beta type gene achieved a validated diagnostic accuracy of 100% in tissue and in circulating free DNA isolated from patient plasma. CONCLUSION The success of machine-learning to identify an effective marker signature documents the power of this approach. The high diagnostic accuracy of discriminating PDAC from CP could have tremendous consequences for treatment success, once the result from still a limited number of liquid biopsy samples would be confirmed in a larger cohort of patients with suspected pancreatic cancer.
Collapse
Affiliation(s)
- Yenan Wu
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Isabelle Seufert
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Fawaz N Al-Shaheri
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Roman Kurilov
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andrea S Bauer
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mehdi Manoochehri
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Evgeny A Moskalev
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christin Tjaden
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Nathalia A Giese
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thilo Hackert
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Markus W Büchler
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Jörg D Hoheisel
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
14
|
Jajodia A, Wang A, Alabousi M, Wilks C, Kulkarni A, van der Pol CB. MRI vs. CT for pancreatic adenocarcinoma vascular invasion: comparative diagnostic test accuracy systematic review and meta-analysis. Eur Radiol 2023; 33:6883-6891. [PMID: 37083741 DOI: 10.1007/s00330-023-09659-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/26/2023] [Accepted: 02/22/2023] [Indexed: 04/22/2023]
Abstract
OBJECTIVES To perform a systematic review comparing the diagnostic accuracy of MRI vs. CT for assessing pancreatic ductal adenocarcinoma (PDAC) vascular invasion. METHODS MEDLINE, EMBASE, Cochrane Central, and Scopus were searched until December 2021 for diagnostic accuracy studies comparing MRI vs. CT to evaluate vascular invasion of pathologically confirmed PDAC in the same patients. Findings on resection or exploratory laparotomy were the preferred reference standard. Data extraction, risk of bias, and applicability assessment were performed by two authors using the Quality Assessment of Diagnostic Accuracy Studies-Comparative Tool. Bivariate random-effects meta-analysis and meta-regression were performed with 95% confidence intervals (95% CI). RESULTS Three studies were included assessing 474 vessels without vascular invasion and 65 with vascular invasion in 107 patients. All patients were imaged using MRI at ≥ 1.5 T and a pancreatic protocol CT. No difference was shown between MRI and CT for diagnosing PDAC vascular invasion: MRI/CT sensitivity (95% CI) were 71% (47-87%)/74% (56-86%), and specificity were 97% (94-99%)/97% (94-98%). Sources of bias included selection bias from only a subset of CT patients undergoing MRI and verification bias from patients with unresectable disease not confirmed on surgery. No patients received neoadjuvant therapy prior to staging. CONCLUSIONS Based on limited data, no difference was observed between MRI and pancreatic protocol CT for PDAC vascular invasion assessment. MRI may be an adequate substitute for pancreatic protocol CT in some patients, particularly those who have already had a single-phase CT. Larger and more recent cohort studies at low risk of bias, including patients who have received neoadjuvant therapy, are needed. CLINICAL RELEVANCE STATEMENT Abdominal MRI performed similarly to pancreatic protocol CT at assessing pancreatic ductal adenocarcinoma vascular invasion, suggesting local staging is adequate in some patients using MRI. More data are needed using larger, more recent cohorts including patients with neoadjuvant treatment. KEY POINTS • Based on limited data, no difference was found between MRI and pancreatic protocol CT sensitivity and specificity for diagnosing PDAC vascular invasion (p = 0.81, 0.73 respectively). • Risk of bias could be reduced in future PDAC MRI vs CT comparative diagnostic test accuracy research by ensuring all enrolled patients undergo both imaging modalities being compared in random order and regardless of the findings on either modality. • More studies are needed that directly compare the diagnostic performance of MRI and CT for PDAC staging after neoadjuvant therapy.
Collapse
Affiliation(s)
- Ankush Jajodia
- Department of Radiology, McMaster University, Hamilton Health Sciences, Hamilton, Canada
| | - Ashley Wang
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Canada
| | - Mostafa Alabousi
- Joint Department of Medical Imaging, University of Toronto, University Health Network, Toronto, Canada
| | - Christopher Wilks
- Department of Radiology, McMaster University, Hamilton Health Sciences, Hamilton, Canada
| | - Ameya Kulkarni
- Department of Radiology, McMaster University, Hamilton Health Sciences, Hamilton, Canada
- Department of Diagnostic Imaging, Juravinski Hospital and Cancer Centre, Hamilton Health Sciences, 711 Concession Street, Hamilton, ON, L8V 1C3, Canada
| | - Christian B van der Pol
- Department of Radiology, McMaster University, Hamilton Health Sciences, Hamilton, Canada.
- Department of Diagnostic Imaging, Juravinski Hospital and Cancer Centre, Hamilton Health Sciences, 711 Concession Street, Hamilton, ON, L8V 1C3, Canada.
| |
Collapse
|
15
|
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: 2.0] [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
|
16
|
Tornel-Avelar AI, Velarde Ruiz-Velasco JA, Pelaez-Luna M. Pancreatic cancer, autoimmune or chronic pancreatitis, beyond tissue diagnosis: Collateral imaging and clinical characteristics may differentiate them. World J Gastrointest Oncol 2023; 15:925-942. [PMID: 37389107 PMCID: PMC10302998 DOI: 10.4251/wjgo.v15.i6.925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/21/2023] [Accepted: 04/28/2023] [Indexed: 06/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies and is developing into the 2nd leading cause of cancer-related death. Often, the clinical and radiological presentation of PDAC may be mirrored by other inflammatory pancreatic masses, such as autoimmune pancreatitis (AIP) and mass-forming chronic pancreatitis (MFCP), making its diagnosis challenging. Differentiating AIP and MFCP from PDAC is vital due to significant therapeutic and prognostic implications. Current diagnostic criteria and tools allow the precise differentiation of benign from malignant masses; however, the diagnostic accuracy is imperfect. Major pancreatic resections have been performed in AIP cases under initial suspicion of PDAC after a diagnostic approach failed to provide an accurate diagnosis. It is not unusual that after a thorough diagnostic evaluation, the clinician is confronted with a pancreatic mass with uncertain diagnosis. In those cases, a re-evaluation must be entertained, preferably by an experienced multispecialty team including radiologists, pathologists, gastroenterologists, and surgeons, looking for disease-specific clinical, imaging, and histological hallmarks or collateral evidence that could favor a specific diagnosis. Our aim is to describe current diagnostic limitations that hinder our ability to reach an accurate diagnosis among AIP, PDAC, and MFCP and to highlight those disease-specific clinical, radiological, serological, and histological characteristics that could support the presence of any of these three disorders when facing a pancreatic mass with uncertain diagnosis after an initial diagnostic approach has been unsuccessful.
Collapse
Affiliation(s)
- Ana I Tornel-Avelar
- Department of Gastroenterology, Hospital Civil of Guadalajara “Fray Antonio Alcalde”, Guadalajara 44340, Jalisco, Mexico
| | | | - Mario Pelaez-Luna
- Research Division School of Medicine/Department of Gastroenterology, Universidad Nacional Autonoma de México/National Institute of Medical Sciences and Nutrition “Salvador Zubiran”, Tlalpan 14000, Mexico City, Mexico
| |
Collapse
|
17
|
Yamada R, Tsuboi J, Murashima Y, Tanaka T, Nose K, Nakagawa H. Advances in the Early Diagnosis of Pancreatic Ductal Adenocarcinoma and Premalignant Pancreatic Lesions. Biomedicines 2023; 11:1687. [PMID: 37371782 DOI: 10.3390/biomedicines11061687] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/23/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Pancreatic cancer is one of the most lethal human malignancies, in part because it is often diagnosed at late stages when surgery and systemic therapies are either unfeasible or ineffective. Therefore, diagnosing pancreatic cancer in earlier stages is important for effective treatment. However, because the signs and symptoms may be nonspecific and not apparent until the disease is at a late stage, the timely diagnoses of pancreatic cancer can be difficult to achieve. Recent studies have shown that selective screening and increased usage of biomarkers could improve the early diagnosis of pancreatic cancer. In this review, we discuss recent advancements in the early detection of pancreatic ductal carcinoma and precancerous lesions. These include innovations in imaging modalities, the diagnostic utility of various biomarkers, biopsy techniques, and population-based surveillance approaches. Additionally, we discuss how machine learning methods are being applied to develop integrated methods of identifying individuals at high risk of developing pancreatic disease. In the future, the overall survival of pancreatic cancer patients could be improved by the development and adoption of these new methods and techniques.
Collapse
Affiliation(s)
- Reiko Yamada
- Department of Gastroenterology and Hepatology, School of Medicine, Mie University, Tsu 514-8507, Japan
| | - Junya Tsuboi
- Department of Gastroenterology and Hepatology, School of Medicine, Mie University, Tsu 514-8507, Japan
| | - Yumi Murashima
- Department of Gastroenterology and Hepatology, School of Medicine, Mie University, Tsu 514-8507, Japan
| | - Takamitsu Tanaka
- Department of Gastroenterology and Hepatology, School of Medicine, Mie University, Tsu 514-8507, Japan
| | - Kenji Nose
- Department of Gastroenterology and Hepatology, School of Medicine, Mie University, Tsu 514-8507, Japan
| | - Hayato Nakagawa
- Department of Gastroenterology and Hepatology, School of Medicine, Mie University, Tsu 514-8507, Japan
| |
Collapse
|
18
|
Ran H, Chen G, He Y, Yu Q, Xie Y, Liu J, Liu H, Zhang T. Undifferentiated carcinoma with osteoclast‑like giant cells of the pancreas: A case report. Oncol Lett 2023; 25:252. [PMID: 37153037 PMCID: PMC10161351 DOI: 10.3892/ol.2023.13838] [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: 01/16/2023] [Accepted: 03/31/2023] [Indexed: 05/09/2023] Open
Abstract
Undifferentiated carcinoma with osteoclast-like giant cells of the pancreas (UCOGCP) is a rare pancreatic tumor that accounts for <1% of all primary pancreatic malignant tumors. Although the tumor is considered a variant of pancreatic ductal adenocarcinoma, there are substantial differences in the clinicopathological characteristics between UCOGCP and pancreatic ductal adenocarcinoma. Imaging examinations are useful in making a correct diagnosis, and providing a reasonable and effective surgical treatment regimen; however, the imaging characteristics of UCOGCP require further investigation. The present report describes a rare case of UCOGCP with rapid progression and poor prognosis. The patient could not undergo surgery and received chemotherapy drugs only. Chemotherapy did not markedly improve the outcome, and a follow-up 6 months after discharge showed that the patient had died. The present report describes this case and summarizes the available imaging findings to increase awareness, and to improve early diagnosis of this rare disease and therapeutic outcomes.
Collapse
Affiliation(s)
- Haifeng Ran
- Department of Radiology, Medical Imaging Center of Guizhou Province, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Guiqin Chen
- Department of Radiology, Medical Imaging Center of Guizhou Province, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yulun He
- Department of Radiology, Medical Imaging Center of Guizhou Province, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Qiane Yu
- Department of Radiology, Medical Imaging Center of Guizhou Province, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yuxin Xie
- Department of Radiology, Medical Imaging Center of Guizhou Province, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Junwei Liu
- Department of Radiology, Medical Imaging Center of Guizhou Province, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Heng Liu
- Department of Radiology, Medical Imaging Center of Guizhou Province, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Correspondence to: Professor Tijiang Zhang or Professor Heng Liu, Department of Radiology, Medical Imaging Center of Guizhou Province, The Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, Guizhou 563000, P.R. China, E-mail:
| | - Tijiang Zhang
- Department of Radiology, Medical Imaging Center of Guizhou Province, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Correspondence to: Professor Tijiang Zhang or Professor Heng Liu, Department of Radiology, Medical Imaging Center of Guizhou Province, The Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, Guizhou 563000, P.R. China, E-mail:
| |
Collapse
|
19
|
Nannan L, Gsell W, Belderbos S, Gallet C, Wouters J, Brassart-Pasco S, Himmelreich U, Brassart B. A multimodal imaging study to highlight elastin-derived peptide pro-tumoral effect in a pancreatic xenograft model. Br J Cancer 2023; 128:2000-2012. [PMID: 37002342 PMCID: PMC10206107 DOI: 10.1038/s41416-023-02242-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 02/28/2023] [Accepted: 03/16/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is highly malignant with a very poor prognosis due to its silent development and metastatic profile with a 5-year survival rate below 10%. PDAC is characterised by an abundant desmoplastic stroma modulation that influences cancer development by extracellular matrix/cell interactions. Elastin is a key element of the extracellular matrix. Elastin degradation products (EDPs) regulate numerous biological processes such as cell proliferation, migration and invasion. The aim of the present study was to characterise for the first time the effect of two EDPs with consensus sequences "GxxPG" and "GxPGxGxG" (VG-6 and AG-9) on PDAC development. The ribosomal protein SA (RPSA) has been discovered recently, acting as a new receptor of EDPs on the surface of tumour cells, contributing to poor prognosis. METHODS Six week-old female Swiss nude nu/nu (Nu(Ico)-Foxn1nu) mice were subcutaneously injected with human PDAC MIA PaCa-2/eGFP-FLuc+ cells, transduced with a purpose-made lentiviral vector, encoding green fluorescent protein (GFP) and Photinus pyralis (firefly) luciferase (FLuc). Animals were treated three times per week with AG-9 (n = 4), VG-6 (n = 5) or PBS (n = 5). The influence of EDP on PDAC was examined by multimodal imaging (bioluminescence imaging (BLI), fluorescence imaging (FLI) and magnetic resonance imaging (MRI). Tumour volumes were also measured using a caliper. Finally, immunohistology was performed at the end of the in vivo study. RESULTS After in vitro validation of MIA PaCa-2 cells by optical imaging, we demonstrated that EDPs exacerbate tumour growth in the PDAC mouse model. While VG-6 stimulated tumour growth to some extent, AG-9 had greater impact on tumour growth. We showed that the expression of the RPSA correlates with a possible effect of EDPs in the PDAC model. Multimodal imaging allowed for longitudinal in vivo follow-up of tumour development. In all groups, we showed mature vessels ending in close vicinity of the tumour, except for the AG-9 group where mature vessels are penetrating the tumour reflecting an increase of vascularisation. CONCLUSIONS Our results suggest that AG-9 strongly increases PDAC progression through an increase in tumour vascularisation.
Collapse
Affiliation(s)
- Lise Nannan
- KU Leuven, Department of Imaging and Pathology/Biomedical MRI, Leuven, Belgium
- CNRS UMR 7369 Matrice Extracellulaire et Dynamique Cellulaire, Reims, France
- Université de Reims Champagne Ardenne, Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France
| | - Willy Gsell
- KU Leuven, Department of Imaging and Pathology/Biomedical MRI, Leuven, Belgium
| | - Sarah Belderbos
- KU Leuven, Department of Imaging and Pathology/Biomedical MRI, Leuven, Belgium
| | - Célia Gallet
- CNRS UMR 7369 Matrice Extracellulaire et Dynamique Cellulaire, Reims, France
- Université de Reims Champagne Ardenne, Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France
| | - Jens Wouters
- KU Leuven, Department of Imaging and Pathology/Biomedical MRI, Leuven, Belgium
| | - Sylvie Brassart-Pasco
- CNRS UMR 7369 Matrice Extracellulaire et Dynamique Cellulaire, Reims, France
- Université de Reims Champagne Ardenne, Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France
| | - Uwe Himmelreich
- KU Leuven, Department of Imaging and Pathology/Biomedical MRI, Leuven, Belgium
| | - Bertrand Brassart
- CNRS UMR 7369 Matrice Extracellulaire et Dynamique Cellulaire, Reims, France.
- Université de Reims Champagne Ardenne, Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.
| |
Collapse
|
20
|
Marin AM, Sanchuki HBS, Namur GN, Uno M, Zanette DL, Aoki MN. Circulating Cell-Free Nucleic Acids as Biomarkers for Diagnosis and Prognosis of Pancreatic Cancer. Biomedicines 2023; 11:biomedicines11041069. [PMID: 37189687 DOI: 10.3390/biomedicines11041069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
A lack of reliable early diagnostic tools represents a major challenge in the management of pancreatic cancer (PCa), as the disease is often only identified after it reaches an advanced stage. This highlights the urgent need to identify biomarkers that can be used for the early detection, staging, treatment monitoring, and prognosis of PCa. A novel approach called liquid biopsy has emerged in recent years, which is a less- or non-invasive procedure since it focuses on plasmatic biomarkers such as DNA and RNA. In the blood of patients with cancer, circulating tumor cells (CTCs) and cell-free nucleic acids (cfNAs) have been identified such as DNA, mRNA, and non-coding RNA (miRNA and lncRNA). The presence of these molecules encouraged researchers to investigate their potential as biomarkers. In this article, we focused on circulating cfNAs as plasmatic biomarkers of PCa and analyzed their advantages compared to traditional biopsy methods.
Collapse
Affiliation(s)
- Anelis Maria Marin
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Prof Algacyr Munhoz Mader 3775 Street, Curitiba 81350-010, Brazil
| | - Heloisa Bruna Soligo Sanchuki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Prof Algacyr Munhoz Mader 3775 Street, Curitiba 81350-010, Brazil
| | - Guilherme Naccache Namur
- Center for Translational Research in Oncology (LIM24), Departamento de Radiologia e Oncologia, Instituto do Câncer do Estado de São Paulo (ICESP), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo 01246-000, Brazil
| | - Miyuki Uno
- Center for Translational Research in Oncology (LIM24), Departamento de Radiologia e Oncologia, Instituto do Câncer do Estado de São Paulo (ICESP), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo 01246-000, Brazil
| | - Dalila Luciola Zanette
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Prof Algacyr Munhoz Mader 3775 Street, Curitiba 81350-010, Brazil
| | - Mateus Nóbrega Aoki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Prof Algacyr Munhoz Mader 3775 Street, Curitiba 81350-010, Brazil
| |
Collapse
|
21
|
Tikhonova VS, Karmazanovsky GG, Kondratyev EV, Gruzdev IS, Mikhaylyuk KA, Sinelnikov MY, Revishvili AS. Radiomics model-based algorithm for preoperative prediction of pancreatic ductal adenocarcinoma grade. Eur Radiol 2023; 33:1152-1161. [PMID: 35986774 DOI: 10.1007/s00330-022-09046-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/24/2022] [Accepted: 07/13/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To develop diagnostic radiomic model-based algorithm for pancreatic ductal adenocarcinoma (PDAC) grade prediction. METHODS Ninety-one patients with histologically confirmed PDAC and preoperative CT were divided into subgroups based on tumor grade. Two histology-blinded radiologists independently segmented lesions for quantitative texture analysis in all contrast enhancement phases. The ratio of densities of PDAC and unchanged pancreatic tissue, and relative tumor enhancement (RTE) in arterial, portal venous, and delayed phases of the examination were calculated. Principal component analysis was used for multivariate predictor analysis. The selection of predictors in the binary logistic model was carried out in 2 stages: (1) using one-factor logistic models (selection criterion was p < 0.1); (2) using regularization (LASSO regression after standardization of variables). Predictors were included in proportional odds models without interactions. RESULTS There were significant differences in 4, 16, and 8 texture features out of 62 for the arterial, portal venous, and delayed phases of the study, respectively (p < 0.1). After selection, the final diagnostic model included such radiomics features as DISCRETIZED HU standard, DISCRETIZED HUQ3, GLCM Correlation, GLZLM LZLGE for the portal venous phase of the contrast enhancement, and CONVENTIONAL_HUQ3 for the delayed phase of CT study. On its basis, a diagnostic model was built, showing AUC for grade ≥ 2 of 0.75 and AUC for grade 3 of 0.66. CONCLUSION Radiomics features vary in PDAC of different grades and increase the accuracy of CT in preoperative diagnosis. We have developed a diagnostic model, including texture features, which can be used to predict the grade of PDAC. KEY POINTS • A diagnostic algorithm based on CT texture features for preoperative PDAC grade prediction was developed. • The assumption that the scanning protocol can influence the results of texture analysis was confirmed and assessed. • Our results show that tumor differentiation grade can be assessed with sufficient diagnostic accuracy using CT texture analysis presented in this study.
Collapse
Affiliation(s)
| | - Grigory G Karmazanovsky
- A.V. Vishnevsky National Medical Research Centre of Surgery, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - Ivan S Gruzdev
- A.V. Vishnevsky National Medical Research Centre of Surgery, Moscow, Russia
| | | | - Mikhail Y Sinelnikov
- Research Institute of Human Morphology, Moscow, Russia.
- Sechenov University, Moscow, Russia.
| | | |
Collapse
|
22
|
Yang P, Mao K, Gao Y, Wang Z, Wang J, Chen Y, Ma C, Bian Y, Shao C, Lu J. Tumor size measurements of pancreatic cancer with neoadjuvant therapy based on RECIST guidelines: is MRI as effective as CT? Cancer Imaging 2023; 23:8. [PMID: 36653861 PMCID: PMC9850516 DOI: 10.1186/s40644-023-00528-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVES To compare tumor size measurements using CT and MRI in pancreatic cancer (PC) patients with neoadjuvant therapy (NAT). METHODS This study included 125 histologically confirmed PC patients who underwent NAT. The tumor sizes from CT and MRI before and after NAT were compared by using Bland-Altman analyses and intraclass correlation coefficients (ICCs). Variations in tumor size estimates between MRI and CT in relationship to different factors, including NAT methods (chemotherapy, chemoradiotherapy), tumor locations (head/neck, body/tail), tumor regression grade (TRG) levels (0-2, 3), N stages (N0, N1/N2) and tumor resection margin status (R0, R1), were further analysed. The McNemar test was used to compare the efficacy of NAT evaluations based on the CT and MRI measurements according to RECIST 1.1 criteria. RESULTS There was no significant difference between the median tumor sizes from CT and MRI before and after NAT (P = 0.44 and 0.39, respectively). There was excellent agreement in tumor size between MRI and CT, with mean size differences and limits of agreement (LOAs) of 1.5 [-9.6 to 12.7] mm and 0.9 [-12.6 to 14.5] mm before NAT (ICC, 0.93) and after NAT (ICC, 0.91), respectively. For all the investigated factors, there was good or excellent correlation (ICC, 0.76 to 0.95) for tumor sizes between CT and MRI. There was no significant difference in the efficacy evaluation of NAT between CT and MRI measurements (P = 1.0). CONCLUSION MRI and CT have similar performance in assessing PC tumor size before and after NAT.
Collapse
Affiliation(s)
- Panpan Yang
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Kuanzheng Mao
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China ,grid.267139.80000 0000 9188 055XSchool of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yisha Gao
- grid.73113.370000 0004 0369 1660Department of Pathology, Changhai Hospital of Shanghai, Naval Medical University, Shanghai, China
| | - Zhen Wang
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Jun Wang
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Yufei Chen
- grid.24516.340000000123704535College of Electronic and Information Engineering, Tongji University, Shanghai, China
| | - Chao Ma
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China ,grid.24516.340000000123704535College of Electronic and Information Engineering, Tongji University, Shanghai, China
| | - Yun Bian
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Chengwei Shao
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Jianping Lu
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| |
Collapse
|
23
|
Granata V, Fusco R, Setola SV, Galdiero R, Maggialetti N, Silvestro L, De Bellis M, Di Girolamo E, Grazzini G, Chiti G, Brunese MC, Belli A, Patrone R, Palaia R, Avallone A, Petrillo A, Izzo F. Risk Assessment and Pancreatic Cancer: Diagnostic Management and Artificial Intelligence. Cancers (Basel) 2023; 15:351. [PMID: 36672301 PMCID: PMC9857317 DOI: 10.3390/cancers15020351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Pancreatic cancer (PC) is one of the deadliest cancers, and it is responsible for a number of deaths almost equal to its incidence. The high mortality rate is correlated with several explanations; the main one is the late disease stage at which the majority of patients are diagnosed. Since surgical resection has been recognised as the only curative treatment, a PC diagnosis at the initial stage is believed the main tool to improve survival. Therefore, patient stratification according to familial and genetic risk and the creation of screening protocol by using minimally invasive diagnostic tools would be appropriate. Pancreatic cystic neoplasms (PCNs) are subsets of lesions which deserve special management to avoid overtreatment. The current PC screening programs are based on the annual employment of magnetic resonance imaging with cholangiopancreatography sequences (MR/MRCP) and/or endoscopic ultrasonography (EUS). For patients unfit for MRI, computed tomography (CT) could be proposed, although CT results in lower detection rates, compared to MRI, for small lesions. The actual major limit is the incapacity to detect and characterize the pancreatic intraepithelial neoplasia (PanIN) by EUS and MR/MRCP. The possibility of utilizing artificial intelligence models to evaluate higher-risk patients could favour the diagnosis of these entities, although more data are needed to support the real utility of these applications in the field of screening. For these motives, it would be appropriate to realize screening programs in research settings.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, 41012 Napoli, Italy
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via della Signora 2, 20122 Milan, Italy
| | - Sergio Venanzio Setola
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Roberta Galdiero
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Nicola Maggialetti
- Department of Medical Science, Neuroscience and Sensory Organs (DSMBNOS), University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Lucrezia Silvestro
- Division of Clinical Experimental Oncology Abdomen, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Mario De Bellis
- Division of Gastroenterology and Digestive Endoscopy, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Elena Di Girolamo
- Division of Gastroenterology and Digestive Endoscopy, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Giulia Grazzini
- Department of Emergency Radiology, University Hospital Careggi, Largo Brambilla 3, 50134 Florence, Italy
| | - Giuditta Chiti
- Department of Emergency Radiology, University Hospital Careggi, Largo Brambilla 3, 50134 Florence, Italy
| | - Maria Chiara Brunese
- Diagnostic Imaging Section, Department of Medical and Surgical Sciences & Neurosciences, University of Molise, 86100 Campobasso, Italy
| | - Andrea Belli
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Renato Patrone
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Raffaele Palaia
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Antonio Avallone
- Division of Clinical Experimental Oncology Abdomen, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Antonella Petrillo
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Francesco Izzo
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| |
Collapse
|
24
|
Li J, Fu C, Zhao S, Pu Y, Yang F, Zeng S, Yang C, Gao H, Chen L. The progress of PET/MRI in clinical management of patients with pancreatic malignant lesions. Front Oncol 2023; 13:920896. [PMID: 37188192 PMCID: PMC10175752 DOI: 10.3389/fonc.2023.920896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Recently, the morbidity and mortality of pancreatic cancer have been increasing year by year. Because of its deep anatomical location and because most presented patients often suffer from abdominal pain or jaundice, it is difficult to diagnose pancreatic cancer at an early stage, leading to late clinical stage and poor prognosis. integrated positron emission tomography/magnetic resonance imaging (PET/MRI) fusion imaging not only has the characteristics of high resolution and multi-parameter imaging of MRI, but also combines the high sensitivity and the semi-quantitative characteristics of PET. In addition, the continuous development of novel MRI imaging and PET imaging biomarkers provide a unique and precise research direction for future pancreatic cancer research. This review summarizes the value of PET/MRI in the diagnosis, staging, efficacy monitoring, and prognosis evaluation of pancreatic cancer, and prognosis for developing emerging imaging agents and artificial intelligence radiomics in pancreatic cancer.
Collapse
Affiliation(s)
- Jindan Li
- Department of PET-CT/MR Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Chaojiang Fu
- Department of Emergency, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Sheng Zhao
- Department of PET-CT/MR Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yongzhu Pu
- Department of PET-CT/MR Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Fake Yang
- Department of PET-CT/MR Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Shuguang Zeng
- Department of Information Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Conghui Yang
- Department of PET-CT/MR Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hongqiang Gao
- Department of Hepatobiliary Surgery, The First People’s Hospital of Kunming City & Ganmei Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Long Chen, ; Hongqiang Gao,
| | - Long Chen
- Department of PET-CT/MR Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
- *Correspondence: Long Chen, ; Hongqiang Gao,
| |
Collapse
|
25
|
Shaikh S. Editorial for "Abbreviated Gadoxetic Acid-Enhanced MRI for the Detection of Liver Metastases in Patients With Potentially Resectable Pancreatic Ductal Adenocarcinoma". J Magn Reson Imaging 2022; 56:737-738. [PMID: 34997663 DOI: 10.1002/jmri.28054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 11/07/2022] Open
Affiliation(s)
- Sikandar Shaikh
- Department of Radiology, Shadan Institute of Medical Sciences, Hyderabad, Telangana, India
| |
Collapse
|
26
|
Karan A, Kiamos A, Stack A, Gharia B. Pancreatic herniation: a large pancreatic mass concealed within the intrathoracic cavity. BMJ Case Rep 2022; 15:e251039. [PMID: 35878967 PMCID: PMC9328086 DOI: 10.1136/bcr-2022-251039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2022] [Indexed: 11/03/2022] Open
Abstract
Cancer of unknown primary is a challenging entity. We present an elderly woman with metastatic cancer of unknown primary despite comprehensive imaging and immunohistochemical analysis. Based on a thorough history, a gastrointestinal source was suspected and a diagnosis of pancreatic cancer concealed within a type IV hiatal hernia was made using multimodal imaging. On review of prior imaging, due to the highly complex anatomy within our patient's hiatal hernia, the pancreatic mass was retroactively noted. While initial imaging may detect metastatic disease, identifying the primary malignancy requires a thorough history and physical examination, multimodal imaging where malignancy is suspected, and immunohistochemical analysis of metastatic deposits. Herniation of pancreatic cancer has not been previously described in the literature and serves as an important reminder of the importance of multimodal imaging in patients with significantly complex anatomy.
Collapse
Affiliation(s)
- Abhinav Karan
- Internal Medicine, University of Florida College of Medicine-Jacksonville, Jacksonville, Florida, USA
| | - Amy Kiamos
- Internal Medicine, University of Florida College of Medicine-Jacksonville, Jacksonville, Florida, USA
| | - Anthony Stack
- Hematology and Oncology, University of Florida College of Medicine-Jacksonville, Jacksonville, Florida, USA
| | - Bharatsinh Gharia
- Hematology and Oncology, University of Florida College of Medicine-Jacksonville, Jacksonville, Florida, USA
| |
Collapse
|
27
|
Nakai A, Kamata K, Hyodo T, Chikugo T, Hara A, Otsuka Y, Tanaka H, Yoshikawa T, Ishikawa R, Okamoto A, Yamazaki T, Omoto S, Minaga K, Yamao K, Takenaka M, Chiba Y, Watanabe T, Matsumoto I, Takeyama Y, Kudo M. Utility of contrast-enhanced harmonic EUS for diagnosis of portal vein invasion by pancreatic cancer. Endosc Ultrasound 2022; 11:401-406. [PMID: 35848657 DOI: 10.4103/eus-d-21-00185] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Background The value of contrast-enhanced harmonic EUS (CH-EUS) for diagnosis of portal vein invasion in patients with pancreatic cancer was evaluated. Patients and Methods This single-center, retrospective study included consecutive patients with pancreatic cancer who underwent both surgical resection after preoperative EUS, CH-EUS, and contrast-enhanced computed tomography (CE-CT) examinations between April 2015 and August 2017. CH-EUS evaluation was performed during the late phase. Portal vein invasion on EUS and CH-EUS was defined as no continuity in the line of the vessel wall. Definition of portal vein invasion on CE-CT was based on the Loyer's criteria. The accuracy of three modalities for diagnosis of invasion into the portal vein was compared using the McNemar's test. Results Eighty-eight patients (mean age: 71.0 years, ratio of male to female: 48:40) were eligible. Postoperative pathological results were as follows: seven cases of portal vein invasion; 81 cases without. Diagnostic accuracy of EUS, CH-EUS, and CE-CT for diagnosing invasion into the portal vein was 72.7%, 93.2%, and 81.8%, respectively. The differences between CH-EUS and CE-CT (P = 0.0094) and CH-EUS and EUS (P = 0.0022) were significant. EUS and CE-CT were comparable. Conclusion CH-EUS is useful for diagnosis of portal vein invasion by pancreatic cancer.
Collapse
Affiliation(s)
- Atsushi Nakai
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Ken Kamata
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Tomoko Hyodo
- Department of Radiology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Takaaki Chikugo
- Department of Diagnostic Pathology, Kindai University Hospital, Osaka-Sayama, Japan
| | - Akane Hara
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Yasuo Otsuka
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Hidekazu Tanaka
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Tomoe Yoshikawa
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Rei Ishikawa
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Ayana Okamoto
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Tomohiro Yamazaki
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Shunsuke Omoto
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kosuke Minaga
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kentaro Yamao
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Mamoru Takenaka
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Yasutaka Chiba
- Clinical Research Center, Kindai University Hospital, Osaka-Sayama, Japan
| | - Tomohiro Watanabe
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Ippei Matsumoto
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Yoshifumi Takeyama
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| |
Collapse
|
28
|
Wu H, Ou S, Zhang H, Huang R, Yu S, Zhao M, Tai S. Advances in biomarkers and techniques for pancreatic cancer diagnosis. Cancer Cell Int 2022; 22:220. [PMID: 35761336 PMCID: PMC9237966 DOI: 10.1186/s12935-022-02640-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 06/18/2022] [Indexed: 12/31/2022] Open
Abstract
Pancreatic cancer is the most lethal type of malignancy and is characterized by high invasiveness without severe symptoms. It is difficult to detect PC at an early stage because of the low diagnostic accuracy of existing routine methods, such as abdominal ultrasound, CT, MRI, and endoscopic ultrasound (EUS). Therefore, it is of value to develop new diagnostic techniques for early detection with high accuracy. In this review, we aim to highlight research progress on novel biomarkers, artificial intelligence, and nanomaterial applications on the diagnostic accuracy of pancreatic cancer.
Collapse
|
29
|
Farrukh J, Balasubramaniam R, James A, Wadhwani SS, Albazaz R. Pancreatic adenocarcinoma: imaging techniques for diagnosis and management. Br J Hosp Med (Lond) 2022; 83:1-12. [PMID: 35653327 DOI: 10.12968/hmed.2022.0065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Pancreatic cancer is a leading cause of death from cancer but only a minority of patients with pancreatic ductal adenocarcinomas are eligible for curative resection. The increasing role of neoadjuvant therapy provides hope of improving outcomes. However, progress is also reliant on advances in imaging that can identify disease earlier and accurately assess treatment response. Computed tomography remains the cornerstone in evaluation of resectability, offering excellent spatial resolution. However, in high-risk patients, additional magnetic resonance imaging and positron emission tomography-computed tomography may further guide treatment decisions. Conventional computed tomography can be limited in its ability to determine disease response after neoadjuvant therapy. Dual-energy computed tomography and computed tomography or magnetic resonance imaging perfusion studies emerging as potentially better alternatives. Combined with pioneering advances in radiomic analysis, these modalities also show promise in analysing tumour heterogeneity and thereby more accurately predicting outcomes. This article reviews these imaging techniques.
Collapse
Affiliation(s)
- Jawaad Farrukh
- Department of Radiology, Royal Stoke University Hospital, University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, UK
| | - Ravivarma Balasubramaniam
- Department of Radiology, Royal Stoke University Hospital, University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, UK
| | - Anitha James
- Department of Radiology, Royal Stoke University Hospital, University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, UK
| | - Sharan S Wadhwani
- Department of Radiology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Raneem Albazaz
- Department of Radiology, St James's University Hospital, The Leeds Teaching Hospitals NHS Trust, Leeds, UK
| |
Collapse
|
30
|
Zhang X, Zeng Z, Liu H, Xu L, Sun X, Xu J, Song G. Recent development of a magneto-optical nanoplatform for multimodality imaging of pancreatic ductal adenocarcinoma. NANOSCALE 2022; 14:3306-3323. [PMID: 35170601 DOI: 10.1039/d1nr08394e] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer. Given its inconspicuous and atypical early symptoms and hidden location, most patients have already reached the terminal stage before diagnosis. At present, the diagnosis of PDAC mainly depends on serological and imaging examinations. However, serum tests cannot identify specific tumor locations and each imaging technology has its own defects, bringing great challenges to the early diagnosis of PDAC. Therefore, it is of great significance to find new strategies for the early and accurate diagnosis of PDAC. In recent years, a magneto-optical nanoplatform integrating near infrared fluorescence, photoacoustic, magnetic resonance imaging, etc. has attracted widespread attention, giving full play to the complementary advantages of each imaging modality. Herein, we summarize the recent advances of imaging modalities in the diagnosis of pancreatic cancer, and then discuss in detail the construction and modification of magneto or/and optical probes for multimodal imaging, and advances in early diagnosis using the combination of various imaging modalities, which can provide potential tools for the early diagnosis or even intraoperative navigation and post-treatment follow-up of PDAC patients.
Collapse
Affiliation(s)
- Xuan Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China.
- Department of Ophthalmology and Otolaryngology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, P. R. China.
| | - Zhiming Zeng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China.
- Department of Ophthalmology and Otolaryngology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, P. R. China.
| | - Huiyi Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China.
| | - Li Xu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China.
| | - Xin Sun
- College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, P. R. China
| | - Jing Xu
- Department of Ophthalmology and Otolaryngology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, P. R. China.
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China.
| |
Collapse
|
31
|
Hoogenboom SA, Engels MML, Chuprin AV, van Hooft JE, LeGout JD, Wallace MB, Bolan CW. Prevalence, features, and explanations of missed and misinterpreted pancreatic cancer on imaging: a matched case-control study. Abdom Radiol (NY) 2022; 47:4160-4172. [PMID: 36127473 PMCID: PMC9626431 DOI: 10.1007/s00261-022-03671-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 01/18/2023]
Abstract
PURPOSE To characterize the prevalence of missed pancreatic masses and pancreatic ductal adenocarcinoma (PDAC)-related findings on CT and MRI between pre-diagnostic patients and healthy individuals. MATERIALS AND METHODS Patients diagnosed with PDAC (2010-2016) were retrospectively reviewed for abdominal CT- or MRI-examinations 1 month-3 years prior to their diagnosis, and subsequently matched to controls in a 1:4 ratio. Two blinded radiologists scored each imaging exam on the presence of a pancreatic mass and secondary features of PDAC. Additionally, original radiology reports were graded based on the revised RADPEER criteria. RESULTS The cohort of 595 PDAC patients contained 60 patients with a pre-diagnostic CT and 27 with an MRI. A pancreatic mass was suspected in hindsight on CT in 51.7% and 50% of cases and in 1.3% and 0.9% of controls by reviewer 1 (p < .001) and reviewer 2 (p < .001), respectively. On MRI, a mass was suspected in 70.4% and 55.6% of cases and 2.9% and 0% of the controls by reviewer 1 (p < .001) and reviewer 2 (p < .001), respectively. Pancreatic duct dilation, duct interruption, focal atrophy, and features of acute pancreatitis is strongly associated with PDAC (p < .001). In cases, a RADPEER-score of 2 or 3 was assigned to 56.3% of the CT-reports and 71.4% of MRI-reports. CONCLUSION Radiological features as pancreatic duct dilation and interruption, and focal atrophy are common first signs of PDAC and are often missed or unrecognized. Further investigation with dedicated pancreas imaging is warranted in patients with PDAC-related radiological findings.
Collapse
Affiliation(s)
- Sanne A. Hoogenboom
- Department of Gastroenterology and Hepatology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224 USA ,Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Megan M. L. Engels
- Department of Gastroenterology and Hepatology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224 USA ,Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Anthony V. Chuprin
- Department of Radiology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224 USA
| | - Jeanin E. van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Jordan D. LeGout
- Department of Radiology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224 USA
| | - Michael B. Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224 USA ,Department of Gastroenterology and Hepatology, Sheikh Shakhbout Medical City, PO Box 11001, Abu Dhabi, UAE ,Khalifa University School of Medicine, PO Box 127788, Abu Dhabi, UAE
| | - Candice W. Bolan
- Department of Radiology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224 USA
| |
Collapse
|
32
|
Evolving pancreatic cancer treatment: From diagnosis to healthcare management. Crit Rev Oncol Hematol 2021; 169:103571. [PMID: 34923121 DOI: 10.1016/j.critrevonc.2021.103571] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
The prognosis of pancreatic ductal adenocarcinoma is still the worst among solid tumors. In this review, a panel of experts addressed the main unanswered questions about the clinical management of this disease, with the aim of providing practical decision support for physicians. On the basis of the evidence available from the literature, the main topics concerning pancreatic cancer are discussed: the diagnosis, as the need for a pathological characterization and the role for germ-line and somatic molecular profiling; the therapeutic management of resectable disease, as the role of upfront surgery or neoadjuvant chemotherapy, the post-operative restaging and the optimal timing foradjuvant chemotherapy, the management of the borderline resectable and locally advanced disease; the metastatic disease and the role of surgery for the management of patients with isolated metastasis and the use of biomarkers of metastatic potential; the role of supportive care and the healthcare management of pancreatic ductal adenocarcinoma.
Collapse
|
33
|
Imaoka H, Sasaki M, Hashimoto Y, Watanabe K, Miyazawa S, Shibuki T, Mitsunaga S, Ikeda M. Impact of Endoscopic Ultrasound-Guided Tissue Acquisition on Decision-Making in Precision Medicine for Pancreatic Cancer: Beyond Diagnosis. Diagnostics (Basel) 2021; 11:1195. [PMID: 34209310 PMCID: PMC8307595 DOI: 10.3390/diagnostics11071195] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
Precision medicine in cancer treatment refers to targeted therapy based on the evaluation of biomarkers. Although precision medicine for pancreatic cancer (PC) remains challenging, novel biomarker-based therapies, such as pembrolizumab, olaparib, and entrectinib, have been emerging. Most commonly, endoscopic ultrasound-guided tissue acquisition (EUS-TA) had been used for the diagnosis of PC until now. However, advances in EUS-TA devices and biomarker testing, especially next-generation sequencing, have opened up the possibility of sequencing of various genes even in limited amounts of tissue samples obtained by EUS-TA, and identifying potential genetic alterations as therapeutic targets. Precision medicine benefits only a small population of patients with PC, but biomarker-based therapy has shown promising results in patients who once had no treatment options. Now, the role of EUS-TA has extended beyond diagnosis into decision-making regarding the treatment of PC. In this review, we mainly discuss tissue sampling by EUS-TA for biomarker testing and the current status of precision medicine for PC.
Collapse
Affiliation(s)
- Hiroshi Imaoka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa 277-8577, Chiba, Japan; (M.S.); (Y.H.); (K.W.); (S.M.); (T.S.); (S.M.); (M.I.)
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Kitano M, Yamashita Y, Kamata K, Ang TL, Imazu H, Ohno E, Hirooka Y, Fusaroli P, Seo DW, Napoléon B, Teoh AYB, Kim TH, Dietrich CF, Wang HP, Kudo M. The Asian Federation of Societies for Ultrasound in Medicine and Biology (AFSUMB) Guidelines for Contrast-Enhanced Endoscopic Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1433-1447. [PMID: 33653627 DOI: 10.1016/j.ultrasmedbio.2021.01.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 01/25/2021] [Accepted: 01/28/2021] [Indexed: 06/12/2023]
Abstract
The Asian Federation of Societies for Ultrasound in Medicine and Biology aimed to provide information on techniques and indications for contrast-enhanced harmonic endoscopic ultrasound (CH-EUS), and to create statements including the level of recommendation. These statements are based on current scientific evidence reviewed by a Consensus Panel of 15 internationally renowned experts. The reliability of clinical questions was measured by agreement rates after voting. Six statements were made on techniques, including suitable contrast agents for CH-EUS, differences between contrast agents, setting of mechanical index, dual imaging and duration and phases for observation. Thirteen statements were made on indications, including pancreatic solid masses, pancreatic cancer staging, pancreatic cystic lesions and mural nodules, detection of subtle pancreatic lesions, gallbladder sludge and polyps, hepatic lesions, lymph nodes, subepithelial lesions, visceral vascular diseases, guidance of fine needle aspiration and evaluation for local therapy. These international expert consensus guidelines will assist endosonographers in conducting CH-EUS according to evidence-based information.
Collapse
Affiliation(s)
- Masayuki Kitano
- Second Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Yasunobu Yamashita
- Second Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| | - Ken Kamata
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Tiing Leong Ang
- Department of Gastroenterology and Hepatology, Changi General Hospital, Singapore
| | - Hiroo Imazu
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Eizaburo Ohno
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School, Nagoya, Japan
| | - Yoshiki Hirooka
- Department of Liver, Biliary Tract and Pancreas Diseases, Fujita Health University, Aichi, Japan
| | - Pietro Fusaroli
- Gastroenterology Unit, Department of Medical and Surgical Sciences, University of Bologna/Hospital of Imola, Imola, Italy
| | - Dong-Wan Seo
- Department of Gastroenterology, Asan Medical Centre, Seoul, Korea
| | - Bertrand Napoléon
- Department of Gastroenterology, Jean Mermoz Private Hospital, Ramsay Generale de Sante, Lyon, France
| | - Anthony Yuen Bun Teoh
- Department of Surgery, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong, China
| | - Tae Hyeon Kim
- Department of Internal Medicine, Wonkwang University College of Medicine, Iksan, South Korea
| | - Christoph F Dietrich
- Department of Internal Medicine (DAIM), Hirslanden Kliniken Beau Site, Salem und Permanence Bern, Switzerland
| | - Hsiu-Po Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| |
Collapse
|
35
|
Chavez JA, Chen W, Freitag CE, Frankel WL. Pancreatic Frozen Section Guides Operative Management With Few Deferrals and Errors. Arch Pathol Lab Med 2021; 146:84-91. [PMID: 33769446 DOI: 10.5858/arpa.2020-0483-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2021] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Pancreatic adenocarcinoma is the third leading cause of cancer death in the United States. Surgery remains the mainstay of treatment, and frozen section analysis is used to confirm diagnosis and determine resectability and margin status. OBJECTIVE.— To evaluate use and accuracy of frozen section and how diagnosis impacts surgical procedure. DESIGN.— We reviewed patients with planned pancreatic resections between January 2014 and March 2019 with at least 1 frozen section. Pathology reports including frozen sections, preoperative cytology, and operative notes were reviewed. Frozen sections were categorized by margin, primary pancreatic diagnosis, metastasis, or vascular resectability. The deferral and error rates and surgeons' response were noted. RESULTS.— We identified 898 planned pancreatic resections and 221 frozen sections that were performed on 152 cases for 102 margins, 94 metastatic lesions, 20 primary diagnoses, and 5 to confirm vascular resectability. The diagnosis was deferred to permanent sections in 13 of 152 cases (8.6%) on 16 of 221 frozen sections (7.2%): 6 for metastasis, 8 for margins, and 2 for primary diagnosis. Discrepancies/errors were identified in 4 of 152 cases (2.6%) and 4 of 221 frozen sections (1.8%). Surgeon's response was different than expected in 8 of 221 frozen sections (3.6%), but their actions were explained by other intraoperative findings in 6 of 8. CONCLUSIONS.— Frozen section remains an important diagnostic tool used primarily for evaluation of margins and metastasis during pancreatectomy. In most cases, a definitive diagnosis is rendered, with occasional deferrals and few errors. Intraoperative findings explain most cases where surgeons act differently than expected based on frozen section diagnosis.
Collapse
Affiliation(s)
- Jesus A Chavez
- From the Department of Pathology, The Ohio State University Wexner Medical Center, Columbus.,Chavez and Chen contributed equally to the work
| | - Wei Chen
- From the Department of Pathology, The Ohio State University Wexner Medical Center, Columbus.,Chavez and Chen contributed equally to the work
| | - C Eric Freitag
- From the Department of Pathology, The Ohio State University Wexner Medical Center, Columbus
| | - Wendy L Frankel
- From the Department of Pathology, The Ohio State University Wexner Medical Center, Columbus
| |
Collapse
|
36
|
González-Gómez R, Pazo-Cid RA, Sarría L, Morcillo MÁ, Schuhmacher AJ. Diagnosis of Pancreatic Ductal Adenocarcinoma by Immuno-Positron Emission Tomography. J Clin Med 2021; 10:1151. [PMID: 33801810 PMCID: PMC8000738 DOI: 10.3390/jcm10061151] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 12/15/2022] Open
Abstract
Diagnosis of pancreatic ductal adenocarcinoma (PDAC) by current imaging techniques is useful and widely used in the clinic but presents several limitations and challenges, especially in small lesions that frequently cause radiological tumors infra-staging, false-positive diagnosis of metastatic tumor recurrence, and common occult micro-metastatic disease. The revolution in cancer multi-"omics" and bioinformatics has uncovered clinically relevant alterations in PDAC that still need to be integrated into patients' clinical management, urging the development of non-invasive imaging techniques against principal biomarkers to assess and incorporate this information into the clinical practice. "Immuno-PET" merges the high target selectivity and specificity of antibodies and engineered fragments toward a given tumor cell surface marker with the high spatial resolution, sensitivity, and quantitative capabilities of positron emission tomography (PET) imaging techniques. In this review, we detail and provide examples of the clinical limitations of current imaging techniques for diagnosing PDAC. Furthermore, we define the different components of immuno-PET and summarize the existing applications of this technique in PDAC. The development of novel immuno-PET methods will make it possible to conduct the non-invasive diagnosis and monitoring of patients over time using in vivo, integrated, quantifiable, 3D, whole body immunohistochemistry working like a "virtual biopsy".
Collapse
Affiliation(s)
- Ruth González-Gómez
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Roberto A. Pazo-Cid
- Medical Oncology Unit, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain;
| | - Luis Sarría
- Digestive Radiology Unit, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain;
| | - Miguel Ángel Morcillo
- Biomedical Application of Radioisotopes and Pharmacokinetics Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain
| | - Alberto J. Schuhmacher
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Fundación Aragonesa para la Investigación y el Desarrollo (ARAID), 50018 Zaragoza, Spain
| |
Collapse
|
37
|
Hwang SH, Park MS. [Radiologic Evaluation for Resectability of Pancreatic Adenocarcinoma]. TAEHAN YONGSANG UIHAKHOE CHI 2021; 82:315-334. [PMID: 36238739 PMCID: PMC9431945 DOI: 10.3348/jksr.2021.0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 11/25/2022]
Abstract
Imaging studies play an important role in the detection, diagnosis, assessment of resectability, staging, and determination of patient-tailored treatment options for pancreatic adenocarcinoma. Recently, for patients diagnosed with borderline resectable or locally advanced pancreatic cancers, it is recommended to consider curative-intent surgery following neoadjuvant or palliative therapy, if possible. This review covers how to interpret imaging tests and what to consider when assessing resectability, diagnosing distant metastasis, and re-assessing the resectability of pancreatic cancer after neoadjuvant or palliative therapy.
Collapse
|
38
|
Jugniot N, Bam R, Meuillet EJ, Unger EC, Paulmurugan R. Current status of targeted microbubbles in diagnostic molecular imaging of pancreatic cancer. Bioeng Transl Med 2021; 6:e10183. [PMID: 33532585 PMCID: PMC7823123 DOI: 10.1002/btm2.10183] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/19/2020] [Accepted: 08/19/2020] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is often associated with a poor prognosis due to silent onset, resistance to therapies, and rapid spreading. Most patients are ineligible for curable surgery as they present with advanced disease at the time of diagnosis. Present diagnostic methods relying on anatomical changes have various limitations including difficulty to discriminate between benign and malignant conditions, invasiveness, the ambiguity of imaging results, or the inability to detect molecular biomarkers of PDAC initiation and progression. Therefore, new imaging technologies with high sensitivity and specificity are critically needed for accurately detecting PDAC and noninvasively characterizing molecular features driving its pathogenesis. Contrast enhanced targeted ultrasound (CETUS) is an upcoming molecular imaging modality that specifically addresses these issues. Unlike anatomical imaging modalities such as CT and MRI, molecular imaging using CETUS is promising for early and accurate detection of PDAC. The use of molecularly targeted microbubbles that bind to neovascular targets can enhance the ultrasound signal specifically from malignant PDAC tissues. This review discusses the current state of diagnostic imaging modalities for pancreatic cancer and places a special focus on ultrasound targeted-microbubble technology together with its clinical translatability for PDAC detection.
Collapse
Affiliation(s)
- Natacha Jugniot
- Department of RadiologyMolecular Imaging Program at Stanford, Stanford UniversityPalo AltoCaliforniaUSA
| | - Rakesh Bam
- Department of RadiologyMolecular Imaging Program at Stanford, Stanford UniversityPalo AltoCaliforniaUSA
| | | | | | - Ramasamy Paulmurugan
- Department of RadiologyMolecular Imaging Program at Stanford, Stanford UniversityPalo AltoCaliforniaUSA
| |
Collapse
|
39
|
Brunner TB, Haustermans K, Huguet F, Morganti AG, Mukherjee S, Belka C, Krempien R, Hawkins MA, Valentini V, Roeder F. ESTRO ACROP guidelines for target volume definition in pancreatic cancer. Radiother Oncol 2021; 154:60-69. [DOI: 10.1016/j.radonc.2020.07.052] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 02/08/2023]
|
40
|
Sahni S, Pandya AR, Hadden WJ, Nahm CB, Maloney S, Cook V, Toft JA, Wilkinson-White L, Gill AJ, Samra JS, Dona A, Mittal A. A unique urinary metabolomic signature for the detection of pancreatic ductal adenocarcinoma. Int J Cancer 2020; 148:1508-1518. [PMID: 33128797 DOI: 10.1002/ijc.33368] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/24/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022]
Abstract
Our study aimed to identify a urinary metabolite panel for the detection/diagnosis of pancreatic ductal adenocarcinoma (PDAC). PDAC continues to have poor survival outcomes. One of the major reasons for poor prognosis is the advanced stage of the disease at diagnosis. Hence, identification of a novel and cost-effective biomarker signature for early detection/diagnosis of PDAC could lead to better survival outcomes. Untargeted metabolomics was employed to identify a novel metabolite-based biomarker signature for PDAC diagnosis. Urinary metabolites from 92 PDAC patients (56 discovery cohort and 36 validation cohort) were compared with 56 healthy volunteers using 1 H nuclear magnetic resonance spectroscopy. Multivariate (partial-least squares discriminate analysis) and univariate (Mann-Whitney's U-test) analyses were performed to identify a metabolite panel which can be used to detect PDAC. The selected metabolites were further validated for their diagnostic potential using the area under the receiver operating characteristic (AUROC) curve. Statistical analysis identified a six-metabolite panel (trigonelline, glycolate, hippurate, creatine, myoinositol and hydroxyacetone), which demonstrated high potential to diagnose PDAC, with AUROC of 0.933 and 0.864 in the discovery and validation cohort, respectively. Notably, the identified panel also demonstrated very high potential to diagnose early-stage (I and II) PDAC patients with AUROC of 0.897. These results demonstrate that the selected metabolite signature could be used to detect PDAC and will pave the way for the development of a urinary test for detection/diagnosis of PDAC.
Collapse
Affiliation(s)
- Sumit Sahni
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia.,Australian Pancreatic Centre, Sydney, New South Wales, Australia
| | - Advait R Pandya
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - William J Hadden
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Christopher B Nahm
- Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia.,Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, New South Wales, Australia
| | - Sarah Maloney
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Victoria Cook
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - James A Toft
- Nepean Clinical School, University of Sydney, New South Wales, Australia
| | | | - Anthony J Gill
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia.,Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Jaswinder S Samra
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Australian Pancreatic Centre, Sydney, New South Wales, Australia.,Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, New South Wales, Australia
| | - Anthony Dona
- Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Anubhav Mittal
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Australian Pancreatic Centre, Sydney, New South Wales, Australia.,Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, New South Wales, Australia
| |
Collapse
|
41
|
Rhee H, Park MS. The Role of Imaging in Current Treatment Strategies for Pancreatic Adenocarcinoma. Korean J Radiol 2020; 22:23-40. [PMID: 32901458 PMCID: PMC7772381 DOI: 10.3348/kjr.2019.0862] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 04/30/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023] Open
Abstract
In pancreatic cancer, imaging plays an essential role in surveillance, diagnosis, resectability evaluation, and treatment response evaluation. Pancreatic cancer surveillance in high-risk individuals has been attempted using endoscopic ultrasound (EUS) or magnetic resonance imaging (MRI). Imaging diagnosis and resectability evaluation are the most important factors influencing treatment decisions, where computed tomography (CT) is the preferred modality. EUS, MRI, and positron emission tomography play a complementary role to CT. Treatment response evaluation is of increasing clinical importance, especially in patients undergoing neoadjuvant therapy. This review aimed to comprehensively review the role of imaging in relation to the current treatment strategy for pancreatic cancer, including surveillance, diagnosis, evaluation of resectability and treatment response, and prediction of prognosis.
Collapse
Affiliation(s)
- Hyungjin Rhee
- 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.
| |
Collapse
|
42
|
Baenas DF, Miretti VS, Caeiro F, Paira S. Differential diagnosis between pancreatic involvement in IgG4-related disease and pancreatic cancer. GASTROENTEROLOGIA Y HEPATOLOGIA 2020; 44:144-155. [PMID: 32718841 DOI: 10.1016/j.gastrohep.2020.05.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/20/2020] [Accepted: 05/20/2020] [Indexed: 12/15/2022]
Abstract
IgG4-related disease is a systemic disorder characterised by diffuse or tumoural inflammatory lesions. It can mimic pancreatic cancer, leading to errors in diagnosis and treatment increasing rates of morbidity and mortality in patients. The aim of this review is to take a differential diagnostic approach to these two entities using epidemiology, clinical and laboratory findings, imaging and histopathology.
Collapse
Affiliation(s)
- Diego Federico Baenas
- Servicio de Reumatología, Hospital Privado Universitario de Córdoba, Córboba, Argentina; Grupo de estudio de enfermedad relacionada con IgG4 de la Sociedad Argentina de Reumatología (SAR), Argentina.
| | - Virginia Soledad Miretti
- Servicio de Oncología y Hematología, Hospital Privado Universitario de Córdoba, Córboba, Argentina
| | - Francisco Caeiro
- Servicio de Reumatología, Hospital Privado Universitario de Córdoba, Córboba, Argentina; Grupo de estudio de enfermedad relacionada con IgG4 de la Sociedad Argentina de Reumatología (SAR), Argentina
| | - Sergio Paira
- Grupo de estudio de enfermedad relacionada con IgG4 de la Sociedad Argentina de Reumatología (SAR), Argentina; Servicio de Reumatología, Hospital J.M. Cullen, Santa Fe, Argentina
| |
Collapse
|
43
|
Diffusion Kurtosis Imaging-A Superior Approach to Assess Tumor-Stroma Ratio in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2020; 12:cancers12061656. [PMID: 32580519 PMCID: PMC7352692 DOI: 10.3390/cancers12061656] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/31/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022] Open
Abstract
Extensive desmoplastic stroma is a hallmark of pancreatic ductal adenocarcinoma (PDAC) and contributes to tumor progression and to the relative resistance of tumor cells towards (radio) chemotherapy. Thus, therapies that target the stroma are under intense investigation. To allow the stratification of patients who would profit from such therapies, non-invasive methods assessing the stroma content in relation to tumor mass are required. In the current prospective study, we investigated the usefulness of diffusion-weighted magnetic resonance imaging (DW-MRI), a radiologic method that measures the random motion of water molecules in tissue, in the assessment of PDAC lesions, and more specifically in the desmoplastic tumor stroma. We made use of a sophisticated DW-MRI approach, the so-called diffusion kurtosis imaging (DKI), which possesses potential advantages over conventional and widely used monoexponential diffusion-weighted imaging analysis (cDWI). We found that the diffusion constant D from DKI is highly negatively correlated with the percentage of tumor stroma, the latter determined by histology. D performed significantly better than the widely used apparent diffusion coefficient (ADC) from cDWI in distinguishing stroma-rich (>50% stroma percentage) from stroma-poor tumors (≤50% stroma percentage). Moreover, we could prove the potential of the diffusion constant D as a clinically useful imaging parameter for the differentiation of PDAC-lesions from non-neoplastic pancreatic parenchyma. Therefore, the diffusion constant D from DKI could represent a valuable non-invasive imaging biomarker for assessment of stroma content in PDAC, which is applicable for the clinical diagnostic of PDAC.
Collapse
|
44
|
Elbanna KY, Jang HJ, Kim TK. Imaging diagnosis and staging of pancreatic ductal adenocarcinoma: a comprehensive review. Insights Imaging 2020; 11:58. [PMID: 32335790 PMCID: PMC7183518 DOI: 10.1186/s13244-020-00861-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 03/06/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has continued to have a poor prognosis for the last few decades in spite of recent advances in different imaging modalities mainly due to difficulty in early diagnosis and aggressive biological behavior. Early PDAC can be missed on CT due to similar attenuation relative to the normal pancreas, small size, or hidden location in the uncinate process. Tumor resectability and its contingency on the vascular invasion most commonly assessed with multi-phasic thin-slice CT is a continuously changing concept, particularly in the era of frequent neoadjuvant therapy. Coexistent celiac artery stenosis may affect the surgical plan in patients undergoing pancreaticoduodenectomy. In this review, we discuss the challenges related to the imaging of PDAC. These include radiological and clinical subtleties of the tumor, evolving imaging criteria for tumor resectability, preoperative diagnosis of accompanying celiac artery stenosis, and post-neoadjuvant therapy imaging. For each category, the key imaging features and potential pitfalls on cross-sectional imaging will be discussed. Also, we will describe the imaging discriminators of potential mimickers of PDAC.
Collapse
Affiliation(s)
- Khaled Y Elbanna
- Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto, Toronto, ON, Canada.
| | - Hyun-Jung Jang
- Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto, Toronto, ON, Canada
| | - Tae Kyoung Kim
- Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
45
|
Pancreatic adenocarcinoma: variability in measurements of tumor size among computed tomography, magnetic resonance imaging, and pathologic specimens. Abdom Radiol (NY) 2020; 45:782-788. [PMID: 31292672 DOI: 10.1007/s00261-019-02125-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE To compare the tumor size measurements assessed by computed tomography (CT) and magnetic resonance imaging (MRI) versus measurements of resected pathologic specimens from patients with pancreatic ductal adenocarcinoma (PDAC). METHODS This study included 114 patients with histologically confirmed PDAC who underwent contrast-enhanced CT and MRI before surgery. The tumor sizes from CT, MRI, and pathologic specimens were compared by using Bland-Altman analyses and intraclass correlation coefficients (ICCs). The discrepancies in PDAC size between CT/MRI and pathologic specimens were calculated and contributing factors for the discrepancies, including tumor locations (pancreatic head/neck, body, or tail), T stages (T1, T2, or T3), and N stages (N0, N1, or N2), were analyzed with Pearson's correlation coefficients and multivariable linear regression analyses. RESULTS There was significant difference among the mean tumor sizes of three measurements (P < 0.001). The difference in mean tumor size between the pathologic sizes for PDAC was 4.3 mm (ICC 0.67) on CT and 5.8 mm (ICC 0.65) on MRI. Both CT and MRI showed wide ranges of limits of agreement (LOAs) between the pathologic specimens for tumor size measurements (LOAs, - 28.9 to 21.4 and - 29.4 to 17.8, respectively). The tumor size on CT or MRI was estimated to be smaller than that on pathology when the tumor was > 30 mm. The discrepancies in the tumor size estimated between CT/MRI and pathologic specimens were significantly different for tumors of different T stages (P < 0.001). CONCLUSIONS Both contrast-enhanced CT and MRI underestimate the mean tumor size by 4.3 mm and 5.8 mm, respectively, compared to the size of pathologic specimens. A larger tumor size indicates a greater discrepancy in the PDAC size measurements between CT/MRI and pathologic specimens.
Collapse
|
46
|
Fernandez Y Viesca M, Arvanitakis M. Early Diagnosis And Management Of Malignant Distal Biliary Obstruction: A Review On Current Recommendations And Guidelines. Clin Exp Gastroenterol 2019; 12:415-432. [PMID: 31807048 PMCID: PMC6842280 DOI: 10.2147/ceg.s195714] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022] Open
Abstract
Malignant biliary obstruction is a challenging condition, requiring a multimodal approach for both diagnosis and treatment. Pancreatic adenocarcinoma and cholangiocarcinoma are the leading causes of malignant distal biliary obstruction. Early diagnosis is difficult to establish as biliary obstruction can be the first presentation of the underlying disease, which can already be at an advanced stage. Consequently, the majority of patients (70%) with malignant distal biliary obstruction are unresectable at the time of diagnosis. The association of clinical findings, laboratory tests, imaging, and endoscopic modalities may help in identifying the underlying cause. Novel endoscopic techniques such as cholangioscopy, intraductal ultrasonography, or confocal laser endomicroscopy have been developed with promising results, but are not used in routine clinical practice. As the number of patients with malignant distal biliary obstruction who will undergo curative surgery is limited, endoscopy has a crucial role in palliation, to relieve biliary obstruction. According to the last European guidelines published in the management of biliary obstruction, self-expandable metal stents have a central place in biliary drainage compared to plastic stents. Endoscopic ultrasound has evolved impressively in the last decades. When standard techniques of biliary cannulation by endoscopic retrograde cholangiopancreatography fail, endoscopic ultrasound-guided biliary drainage is a good option compared to percutaneous drainage.
Collapse
Affiliation(s)
- Michael Fernandez Y Viesca
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Erasme Hospital, Univertié Libre de Bruxelles (ULB), Brussels, Belgium
| | - Marianna Arvanitakis
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Erasme Hospital, Univertié Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
47
|
Lohrmann C, O'Reilly EM, O'Donoghue JA, Pandit-Taskar N, Carrasquillo JA, Lyashchenko SK, Ruan S, Teng R, Scholz W, Maffuid PW, Lewis JS, Weber WA. Retooling a Blood-Based Biomarker: Phase I Assessment of the High-Affinity CA19-9 Antibody HuMab-5B1 for Immuno-PET Imaging of Pancreatic Cancer. Clin Cancer Res 2019; 25:7014-7023. [PMID: 31540979 DOI: 10.1158/1078-0432.ccr-18-3667] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 05/14/2019] [Accepted: 09/06/2019] [Indexed: 01/16/2023]
Abstract
PURPOSE In patients with cancer who have an abnormal biomarker finding, the source of the biomarker in the bloodstream must be located for confirmation of diagnosis, staging, and therapy planning. We evaluated if immuno-PET with the radiolabeled high-affinity antibody HuMab-5B1 (MVT-2163), binding to the cancer antigen CA19-9, can identify the source of elevated biomarkers in patients with pancreatic cancer. PATIENTS AND METHODS In this phase I dose-escalating study, 12 patients with CA19-9-positive metastatic malignancies were injected with MVT-2163. Within 7 days, all patients underwent a total of four whole-body PET/CT scans. A diagnostic CT scan was performed prior to injection of MVT-2163 to correlate findings on MVT-2163 PET/CT. RESULTS Immuno-PET with MVT-2163 was safe and visualized known primary tumors and metastases with high contrast. In addition, radiotracer uptake was not only observed in metastases known from conventional CT, but also seen in subcentimeter lymph nodes located in typical metastatic sites of pancreatic cancer, which were not abnormal on routine clinical imaging studies. A significant fraction of the patients demonstrated very high and, over time, increased uptake of MVT-2163 in tumor tissue, suggesting that HuMab-5B1 labeled with beta-emitting radioisotopes may have the potential to deliver therapeutic doses of radiation to cancer cells. CONCLUSIONS Our study shows that the tumor antigen CA19-9 secreted to the circulation can be used for sensitive detection of primary tumors and metastatic disease by immuno-PET. This significantly broadens the number of molecular targets that can be used for PET imaging and offers new opportunities for noninvasive characterization of tumors in patients.
Collapse
Affiliation(s)
- Christian Lohrmann
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York. .,Weill Cornell Medical College, New York, New York
| | - Eileen M O'Reilly
- Weill Cornell Medical College, New York, New York.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joseph A O'Donoghue
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neeta Pandit-Taskar
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Jorge A Carrasquillo
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Serge K Lyashchenko
- Weill Cornell Medical College, New York, New York.,Radiochemistry and Imaging Sciences Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Shutian Ruan
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rebecca Teng
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Paul W Maffuid
- MabVax Therapeutics Holdings, Inc. San Diego, California
| | - Jason S Lewis
- Weill Cornell Medical College, New York, New York.,Radiochemistry and Imaging Sciences Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Wolfgang A Weber
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
48
|
Wartski M, Sauvanet A. 18F-FDG PET/CT in pancreatic adenocarcinoma: A role at initial imaging staging? Diagn Interv Imaging 2019; 100:735-741. [PMID: 31402332 DOI: 10.1016/j.diii.2019.07.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/05/2019] [Accepted: 07/23/2019] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma represents 90% of all pancreatic tumors. The only hope for prolonged survival in patients with this condition still remains surgery with complete R0 resection. Initial imaging has a pivotal role to identify patients who are eligible to curative surgery and those who may benefit of neoadjuvant chemotherapy. This review provides an analysis of the recent literature on 18F-fluorodeoxyglucose positron emission tomography/computed tomography (FDG PET/CT) in pancreatic adenocarcinoma. Performances of FDG PET in the detection of lymph node involvement and metastatic spread at initial staging and those in the assessment of response to treatment are described.
Collapse
Affiliation(s)
- M Wartski
- Department of Nuclear Medicine, Université de Paris - Paris Descartes, Cochin Hospital-AP-HP, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France.
| | - A Sauvanet
- Department of Hepato-Pancreato-Biliary Surgery, Pôle des Maladies de l'Appareil Digestif, Université de Paris-Paris Diderot, Beaujon Hospital, 92110 Clichy, France
| |
Collapse
|
49
|
Chen Y, Jiang Z, Long L, Miu Y, Zhang L, Zhong D, Tang Q. Magnetic resonance imaging: Proton density fat fraction for assessment of pancreatic fatty infiltration during progression of T2DM bama minipigs. J Magn Reson Imaging 2019; 50:1905-1913. [PMID: 31006935 DOI: 10.1002/jmri.26754] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 04/03/2019] [Accepted: 04/03/2019] [Indexed: 12/15/2022] Open
Affiliation(s)
- Yidi Chen
- First Affiliated Hospital of Guangxi Medical UniversityRadiology Department Guangxi China
| | - Zijian Jiang
- First Affiliated Hospital of Guangxi Medical UniversityRadiology Department Guangxi China
| | - Liling Long
- First Affiliated Hospital of Guangxi Medical UniversityRadiology Department Guangxi China
| | - Yongjian Miu
- First Affiliated Hospital of Guangxi Medical UniversityPathology Department Guangxi China
| | - Ling Zhang
- First Affiliated Hospital of Guangxi Medical UniversityRadiology Department Guangxi China
| | - Delin Zhong
- First Affiliated Hospital of Guangxi Medical UniversityRadiology Department Guangxi China
| | - Qin Tang
- First Affiliated Hospital of Guangxi Medical UniversityRadiology Department Guangxi China
| |
Collapse
|
50
|
Diagnostic and therapeutic recommendations in pancreatic ductal adenocarcinoma. Recommendations of the Working Group of the Polish Pancreatic Club. GASTROENTEROLOGY REVIEW 2019; 14:1-18. [PMID: 30944673 PMCID: PMC6444110 DOI: 10.5114/pg.2019.83422] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 02/15/2019] [Indexed: 02/07/2023]
Abstract
These recommendations refer to the current management in pancreatic ductal adenocarcinoma (PDAC), a neoplasia characterised by an aggressive course and extremely poor prognosis. The recommendations regard diagnosis, surgical, adjuvant and palliative treatment, with consideration given to endoscopic and surgical methods. A vast majority of the statements are based on data obtained in clinical studies and experts' recommendations on PDAC management, including the following guidelines: International Association of Pancreatology/European Pancreatic Club (IAP/EPC), American Society of Clinical Oncology (ASCO), European Society for Medical Oncology (ESMO), National Comprehensive Cancer Network (NCCN) and Polish Society of Gastroenterology (PSG) and The National Institute for Health and Care Excellence (NICE). All recommendations were voted on by members of the Working Group of the Polish Pancreatic Club. Results of the voting and brief comments are provided with each recommendation.
Collapse
|