1
|
Pang X, Zhao Y, Chen X, Wang M, Chen X, Yuan H, Sun Y, Han J, Zhao X. Preclinical Evaluation and First-in-Human Study of [ 68Ga]Ga-αvβ6-2: A Novel Dimeric Integrin αvβ6-Targeted PET Probe for Pancreatic Cancer Imaging. Mol Pharm 2025; 22:2650-2659. [PMID: 40193102 DOI: 10.1021/acs.molpharmaceut.5c00051] [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: 04/24/2025]
Abstract
Integrin αvβ6 is highly expressed in pancreatic cancer, making it an ideal target for molecular imaging diagnosis. Multimerization is considered an effective strategy to increase the accumulation of molecular probes in tumors. Here, we synthesized monomeric and dimeric αvβ6-targeting molecular probes, labeled with 68Ga, and designated them [68Ga]Ga-αvβ6-1 and [68Ga]Ga-αvβ6-2, respectively. Both in vitro and in vivo studies were conducted using human pancreatic cancer BxPC-3 cells and BxPC-3 tumor-bearing mice. Additionally, positron emission tomography/computed tomography (PET/CT) imaging with [68Ga]Ga-αvβ6-2 was performed in three patients with pancreatic cancer. In vitro studies demonstrated that [68Ga]Ga-αvβ6-2 exhibited greater binding affinity, cellular uptake, and internalization than did [68Ga]Ga-αvβ6-1. Micro-PET/CT imaging and biodistribution studies revealed the superior imaging performance of [68Ga]Ga-αvβ6-2. Furthermore, the first-in-human evaluation highlighted the favorable in vivo distribution and diagnostic efficacy of [68Ga]Ga-αvβ6-2 in pancreatic cancer. These results underscore the effectiveness of the multimerization strategy in the application of αvβ6-targeted molecular probes, suggesting that [68Ga]Ga-αvβ6-2 may possess favorable clinical translation potential.
Collapse
Affiliation(s)
- Xiao Pang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Medical Imaging College, North Sichuan Medical College; Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Yan Zhao
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Xiaolin Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Mengjiao Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Xiaoshan Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Huiqing Yuan
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Yuhan Sun
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Jingya Han
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei 050011, China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei 050011, China
| |
Collapse
|
2
|
Piper TB, Schaebel GH, Egeland C, Achiam MP, Burgdorf SK, Nerup N. Fluorescence-guided pancreatic surgery: A scoping review. Surgery 2025; 178:108931. [PMID: 39613658 DOI: 10.1016/j.surg.2024.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 09/05/2024] [Accepted: 10/15/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Although fluorescence guidance during various surgical procedures has been shown to be safe and have possible better clinical outcomes than without the guidance, the use of fluorophores in pancreatic surgery is novel and not yet well described. This scoping review involved a systematic methodology of the currently available literature and aimed to illuminate the use of fluorophores in pancreatic surgery from a clinical view. METHODS The PRISMA and the PRISMA-ScR guidelines were used when appropriate and the following databases were searched: PubMed, Embase, Scopus, The Cochrane Collection, and Web of Science. Human original articles and case reports were included. Bias was assessed with the Newcastle-Ottawa Scale and the IDEAL framework was used for evaluation of surgical innovation. RESULTS A total of 5,565 search hits were screened, and 23 original articles and 24 case reports consisting of 754 patients met the inclusion criteria. The use of indocyanine green was both the most prominent and the most promising method for securing sufficient perfusion of neighboring organs, enhancing the detection and distinguishing of neuroendocrine tumors, and assisting in the identification of hepatic micrometastases. CONCLUSION The included studies were generally heterogenic, exploratory, and small. Indocyanine green was used in several ways, and it may add clinical value in different settings during pancreatic surgery. Tumor-targeted probes are a rapidly developing and promising field of research.
Collapse
Affiliation(s)
- Thomas B Piper
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | - Gustav H Schaebel
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Charlotte Egeland
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark. https://www.twitter.com/ChEgeland
| | - Michael P Achiam
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark. https://www.twitter.com/MichaelAchiam
| | - Stefan K Burgdorf
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Nikolaj Nerup
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark. https://www.twitter.com/nikolajnerup
| |
Collapse
|
3
|
Jafari SH, Lajevardi ZS, Zamani Fard MM, Jafari A, Naghavi S, Ravaei F, Taghavi SP, Mosadeghi K, Zarepour F, Mahjoubin-Tehran M, Rahimian N, Mirzaei H. Imaging Techniques and Biochemical Biomarkers: New Insights into Diagnosis of Pancreatic Cancer. Cell Biochem Biophys 2024; 82:3123-3144. [PMID: 39026059 DOI: 10.1007/s12013-024-01437-z] [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: 07/12/2024] [Indexed: 07/20/2024]
Abstract
Pancreatic cancer (PaC) incidence is increasing, but our current screening and diagnostic strategies are not very effective. However, screening could be helpful in the case of PaC, as recent evidence shows that the disease progresses gradually. Unfortunately, there is no ideal screening method or program for detecting PaC in its early stages. Conventional imaging techniques, such as abdominal ultrasound, CT, MRI, and EUS, have not been successful in detecting early-stage PaC. On the other hand, biomarkers may be a more effective screening tool for PaC and have greater potential for further evaluation compared to imaging. Recent studies on biomarkers and artificial intelligence (AI)-enhanced imaging have shown promising results in the early diagnosis of PaC. In addition to proteins, non-coding RNAs are also being studied as potential biomarkers for PaC. This review consolidates the current literature on PaC screening modalities to provide an organized framework for future studies. While conventional imaging techniques have not been effective in detecting early-stage PaC, biomarkers and AI-enhanced imaging are promising avenues of research. Further studies on the use of biomarkers, particularly non-coding RNAs, in combination with imaging modalities may improve the accuracy of PaC screening and lead to earlier detection of this deadly disease.
Collapse
Affiliation(s)
- Seyed Hamed Jafari
- Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Radiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Sadat Lajevardi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Masoud Zamani Fard
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ameneh Jafari
- Chronic Respiratory Diseases Research Center, NRITLD, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soroush Naghavi
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ravaei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Kimia Mosadeghi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Zarepour
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran; Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
4
|
Saúde-Conde R, El Ghali B, Navez J, Bouchart C, Van Laethem JL. Total Neoadjuvant Therapy in Localized Pancreatic Cancer: Is More Better? Cancers (Basel) 2024; 16:2423. [PMID: 39001485 PMCID: PMC11240662 DOI: 10.3390/cancers16132423] [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: 05/15/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge in oncology due to its advanced stage upon diagnosis and limited treatment options. Surgical resection, the primary curative approach, often results in poor long-term survival rates, leading to the exploration of alternative strategies like neoadjuvant therapy (NAT) and total neoadjuvant therapy (TNT). While NAT aims to enhance resectability and overall survival, there appears to be potential for improvement, prompting consideration of alternative neoadjuvant strategies integrating full-dose chemotherapy (CT) and radiotherapy (RT) in TNT approaches. TNT integrates chemotherapy and radiotherapy prior to surgery, potentially improving margin-negative resection rates and enabling curative resection for locally advanced cases. The lingering question: is more always better? This article categorizes TNT strategies into six main groups based on radiotherapy (RT) techniques: (1) conventional chemoradiotherapy (CRT), (2) the Dutch PREOPANC approach, (3) hypofractionated ablative intensity-modulated radiotherapy (HFA-IMRT), and stereotactic body radiotherapy (SBRT) techniques, which further divide into (4) non-ablative SBRT, (5) nearly ablative SBRT, and (6) adaptive ablative SBRT. A comprehensive analysis of the literature on TNT is provided for both borderline resectable pancreatic cancer (BRPC) and locally advanced pancreatic cancer (LAPC), with detailed sections for each.
Collapse
Affiliation(s)
- Rita Saúde-Conde
- Digestive Oncology Department, Hôpitaux Universitaires de Bruxelles (HUB), Université Libre de Bruxelles, 1070 Brussels, Belgium;
| | - Benjelloun El Ghali
- Department of Radiation Oncology, Hôpitaux Universitaires de Bruxelles (HUB), Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (B.E.G.); (C.B.)
| | - Julie Navez
- Department of Abdominal Surgery and Transplantation, Hôpitaux Universitaires de Bruxelles (HUB), Hopital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium;
| | - Christelle Bouchart
- Department of Radiation Oncology, Hôpitaux Universitaires de Bruxelles (HUB), Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (B.E.G.); (C.B.)
| | - Jean-Luc Van Laethem
- Digestive Oncology Department, Hôpitaux Universitaires de Bruxelles (HUB), Université Libre de Bruxelles, 1070 Brussels, Belgium;
| |
Collapse
|
5
|
Kim BG, Lee SH, Jang Y, Kang S, Kang CM, Cho NH. Differentially expressed genes associated with high metabolic tumor volume served as diagnostic markers and potential therapeutic targets for pancreatic cancer. J Transl Med 2024; 22:453. [PMID: 38741142 PMCID: PMC11092202 DOI: 10.1186/s12967-024-05181-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/05/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND The lack of distinct biomarkers for pancreatic cancer is a major cause of early-stage detection difficulty. The pancreatic cancer patient group with high metabolic tumor volume (MTV), one of the values measured from positron emission tomography-a confirmatory method and standard care for pancreatic cancer, showed a poorer prognosis than those with low MTV. Therefore, MTV-associated differentially expressed genes (DEGs) may be candidates for distinctive markers for pancreatic cancer. This study aimed to evaluate the possibility of MTV-related DEGs as markers or therapeutic targets for pancreatic cancer. METHODS Tumor tissues and their normal counterparts were obtained from patients undergoing preoperative 18F-FDG PET/CT. The tissues were classified into MTV-low and MTV-high groups (7 for each) based on the MTV2.5 value of 4.5 (MTV-low: MTV2.5 < 4.5, MTV-high: MTV2.5 ≥ 4.5). Gene expression fold change was first calculated in cancer tissue compared to its normal counter and then compared between low and high MTV groups to obtain significant DEGs. To assess the suitability of the DEGs for clinical application, the correlation of the DEGs with tumor grades and clinical outcomes was analyzed in TCGA-PAAD, a large dataset without MTV information. RESULTS Total RNA-sequencing (MTV RNA-Seq) revealed that 44 genes were upregulated and 56 were downregulated in the high MTV group. We selected the 29 genes matching MTV RNA-seq patterns in the TCGA-PAAD dataset, a large clinical dataset without MTV information, as MTV-associated genes (MAGs). In the analysis with the TCGA dataset, MAGs were significantly associated with patient survival, treatment outcomes, TCGA-PAAD-suggested markers, and CEACAM family proteins. Some MAGs showed an inverse correlation with miRNAs and were confirmed to be differentially expressed between normal and cancerous pancreatic tissues. Overexpression of KIF11 and RCC1 and underexpression of ADCY1 and SDK1 were detected in ~ 60% of grade 2 pancreatic cancer patients and associated with ~ 60% mortality in stages I and II. CONCLUSIONS MAGs may serve as diagnostic markers and miRNA therapeutic targets for pancreatic cancer. Among the MAGs, KIF11, RCC1, ADCY, and SDK1 may be early diagnostic markers.
Collapse
Affiliation(s)
- Baek Gil Kim
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Hwan Lee
- Division of Hepatobiliary and Pancreas, Department of Surgery, CHA Bundang Medical Center, CHA University, Pocheon, South Korea
| | - Yeonsue Jang
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Suki Kang
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Chang Moo Kang
- Department of Hepatobiliary and Pancreatic Surgery, Yonsei University College of Medicine, Seoul, South Korea.
- Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Seoul, South Korea.
| | - Nam Hoon Cho
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea.
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
6
|
Kandathil A, Subramaniam R. Quarter-Century PET/Computed Tomography Transformation of Oncology: Hepatobiliary and Pancreatic Cancer. PET Clin 2024; 19:163-175. [PMID: 38212214 DOI: 10.1016/j.cpet.2023.12.003] [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: 01/13/2024]
Abstract
[18F] Fluorodeoxyglucose (18F-FDG) PET/CT can improve the staging accuracy and clinical management of patients with hepatobiliary and pancreatic cancers, by detection of unsuspected metastases. 18F-FDG PET/CT metabolic parameters are valuable in predicting treatment response and survival. Metabolic response on 18F-FDG PET/CT can predict preoperative pathologic response to neoadjuvant therapy in patients with pancreatic cancer and determine prognosis. Several novel non-FDG tracers, such as 68Ga prostate-specific membrane antigen (PSMA) and 68Ga-fibroblast activation protein inhibitor (FAPI) PET/CT, show promise for imaging hepatobiliary and pancreatic cancers with potential for radioligand therapy.
Collapse
Affiliation(s)
- Asha Kandathil
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Rathan Subramaniam
- Faculty of Medicine, Nursing, Midwifery and Health Sciences, University of Notre Dame Australia, Sydney, Australia; Department of Radiology, Duke University, Durham, NC, USA; Department of Medicine, University of Otago Medical School, Dunedin, New Zealand
| |
Collapse
|
7
|
Li X, Lu N, Lin L, Chen Y, Yang S, Wang H, Liu X, Wu C, Xue X, Su X, Bai X, Liang T. 18F-FAPI-04 Outperforms 18F-FDG PET/CT in Clinical Assessments of Patients with Pancreatic Adenocarcinoma. J Nucl Med 2024; 65:206-212. [PMID: 38176719 PMCID: PMC10858376 DOI: 10.2967/jnumed.123.266283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/07/2023] [Indexed: 01/06/2024] Open
Abstract
Accurate diagnosis and staging are crucial for selecting treatment for patients with pancreatic ductal adenocarcinoma (PDAC). The desmoplastic responses associated with PDAC are often characterized by hypometabolism. Here, we investigated 18F-fibroblast activation protein inhibitor (FAPI)-04 PET/CT in evaluation of PDAC and compared the findings with those obtained using 18F-FDG. Methods: Sixty-two PDAC patients underwent 18F-FAPI-04 PET/CT and 18F-FDG PET/CT. Identification of primary lesions, lymph node (LN) metastasis, and distant metastasis (DM) by these methods was evaluated, and TNM staging was performed. Correlation between SUVmax of the primary lesion and treatment response was explored in patients who received systemic therapy. Results: 18F-FAPI-04 PET/CT identified all patients with PDAC; 18F-FDG PET/CT missed 1 patient. Tracer uptake was higher in 18F-FAPI-04 PET/CT than in 18F-FDG PET/CT in primary tumors (10.63 vs. 2.87, P < 0.0001), LN metastasis (2.90 vs. 1.43, P < 0.0001), and DM (liver, 6.11 vs. 3.10, P = 0.002; peritoneal, 4.70 vs. 2.08, P = 0.015). The methods showed no significant difference in the T staging category, but the N and M values were significantly higher for 18F-FAPI-04 PET/CT than for 18F-FDG PET/CT (P = 0.002 and 0.008, respectively). Thus, 14 patients were upgraded, and only 1 patient was downgraded, by 18F-FAPI-04 PET/CT compared with 18F-FDG PET/CT. A high SUVmax of the primary tumor did not correlate with treatment response for either 18F-FAPI-04 or 18F-FDG. Conclusion: 18F-FAPI-04 PET/CT performed better than 18F-FDG PET/CT in identification of primary tumors, LN metastasis, and DM and in TNM staging of PDAC.
Collapse
Affiliation(s)
- Xiang Li
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Na Lu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Lili Lin
- Department of Nuclear Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yiwen Chen
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Shuye Yang
- Department of Nuclear Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Huatao Wang
- Department of Nuclear Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinyuan Liu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Chengyi Wu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Xing Xue
- Department of Radiology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; and
| | - Xinhui Su
- Department of Nuclear Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China;
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China;
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
- Zhejiang University Cancer Center, Hangzhou, China
| |
Collapse
|
8
|
Decker JA, Becker J, Härting M, Jehs B, Risch F, Canalini L, Wollny C, Scheurig-Muenkler C, Kroencke T, Schwarz F, Bette S. Optimal conspicuity of pancreatic ductal adenocarcinoma in virtual monochromatic imaging reconstructions on a photon-counting detector CT: comparison to conventional MDCT. Abdom Radiol (NY) 2024; 49:103-116. [PMID: 37796327 PMCID: PMC10789688 DOI: 10.1007/s00261-023-04042-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 10/06/2023]
Abstract
PURPOSE To analyze the conspicuity of pancreatic ductal adenocarcinoma (PDAC) in virtual monoenergetic images (VMI) on a novel photon-counting detector CT (PCD-CT) in comparison to energy-integrating CT (EID-CT). METHODS Inclusion criteria comprised initial diagnosis of PDAC (reference standard: histopathological analysis) and standardized contrast-enhanced CT imaging either on an EID-CT or a PCD-CT. Patients were excluded due to different histopathological diagnosis or missing tumor delineation on CT. On the PCD-CT, 40-190 keV VMI reconstructions were generated. Image noise, tumor-to-pancreas ratio (TPR) and contrast-to-noise ratio (CNR) were analyzed by ROI-based measurements in arterial and portal venous contrast phase. Two board-certified radiologist evaluated image quality and tumor delineation at both, EID-CT and PCD-CT (40 and 70 keV). RESULTS Thirty-eight patients (mean age 70.4 years ± 10.3 [range 45-91], 27 males; PCD-CT: n=19, EID-CT: n=19) were retrospectively included. On the PCD-CT, tumor conspicuity (reflected by low TPR and high CNR) was significantly improved at low-energy VMI series (≤ 70 keV compared to > 70 keV), both in arterial and in portal venous contrast phase (P < 0.001), reaching the maximum at 40 keV. Comparison between PCD-CT and EID-CT showed significantly higher CNR on the PCD-CT in portal venous contrast phase at < 70 keV (P < 0.016). On the PCD-CT, tumor conspicuity was improved in portal venous contrast phase compared to arterial contrast phase especially at the lower end of the VMI spectrum (≤ 70 keV). Qualitative analysis revealed that tumor delineation is improved in 40 keV reconstructions compared to 70 keV reconstructions on a PCD-CT. CONCLUSION PCD-CT VMI reconstructions (≤ 70 keV) showed significantly improved conspicuity of PDAC in quantitative and qualitative analysis in both, arterial and portal venous contrast phase, compared to EID-CT, which may be important for early detection of tumor tissue in clinical routine. Tumor delineation was superior in portal venous contrast phase compared to arterial contrast phase.
Collapse
Affiliation(s)
- Josua A Decker
- Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Augsburg, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany
| | - Judith Becker
- Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Augsburg, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany
| | - Mark Härting
- Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Augsburg, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany
| | - Bertram Jehs
- Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Augsburg, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany
| | - Franka Risch
- Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Augsburg, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany
| | - Luca Canalini
- Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Augsburg, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany
| | - Claudia Wollny
- Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Augsburg, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany
| | - Christian Scheurig-Muenkler
- Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Augsburg, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany
| | - Thomas Kroencke
- Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Augsburg, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany.
- Centre for Advanced Analytics and Predictive Sciences (CAAPS), University of Augsburg, Universitätsstr. 2, 86159, Augsburg, Germany.
| | - Florian Schwarz
- Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Augsburg, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany
- Medical Faculty, Ludwig Maximilian University Munich, Bavariaring 19, 80336, Munich, Germany
- Institute for Radiology, DONAUISAR Hospital Deggendorf-Dingolfing-Landau, Perlasberger Str. 41, 94469, Deggendorf, Germany
| | - Stefanie Bette
- Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Augsburg, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany
| |
Collapse
|
9
|
Duncan ZN, Summerlin D, West JT, Packard AT, Morgan DE, Galgano SJ. PET/MRI for evaluation of patients with pancreatic cancer. Abdom Radiol (NY) 2023; 48:3601-3609. [PMID: 37191756 DOI: 10.1007/s00261-023-03943-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/17/2023]
Abstract
Pancreatic cancers are the third leading cause of cancer-related death in the USA and outcomes remain poor despite improvements in imaging and treatment paradigms. Currently, computed tomography (CT) and magnetic resonance imaging (MRI) are frequently utilized for staging and restaging of these malignancies, but positron emission tomography (PET)/CT can play a role in troubleshooting and improve whole-body staging. PET/MRI is a novel imaging modality that allows for simultaneous acquisition of PET and MRI images, leading to improved image quality and potential increased sensitivity. Early studies suggest that PET/MRI may play a larger role in pancreatic cancer imaging in future. This manuscript will briefly discuss current imaging approaches to pancreatic cancer and outline existing evidence and published data supporting the use of PET/MRI for pancreatic cancers.
Collapse
Affiliation(s)
- Zoey N Duncan
- University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - David Summerlin
- Department of Radiology, University of Alabama at Birmingham Medical Center, Birmingham, AL, USA
| | - Janelle T West
- Department of Radiology, University of Alabama at Birmingham Medical Center, Birmingham, AL, USA
| | | | - Desiree E Morgan
- Department of Radiology, University of Alabama at Birmingham Medical Center, Birmingham, AL, USA
| | - Samuel J Galgano
- Department of Radiology, University of Alabama at Birmingham Medical Center, Birmingham, AL, USA.
| |
Collapse
|
10
|
LeBlanc M, Kang J, Costa AF. Can we rely on contrast-enhanced CT to identify pancreatic ductal adenocarcinoma? A population-based study in sensitivity and factors associated with false negatives. Eur Radiol 2023; 33:7656-7664. [PMID: 37266655 DOI: 10.1007/s00330-023-09758-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/20/2023] [Accepted: 03/27/2023] [Indexed: 06/03/2023]
Abstract
OBJECTIVES To determine the sensitivity of contrast-enhanced computed tomography (CECT) in detecting pancreatic ductal adenocarcinoma (PDAC) and identify factors associated with false negatives (FNs). METHODS Patients diagnosed with PDAC in 2014-2015 were retrospectively identified by a cancer registry. CECTs performed during the diagnostic interval were retrospectively classified as true positive (TP), indeterminate, or FN. Sensitivity TP/(TP+FN) was calculated for all CECTs and the following subgroups: protocol (uniphasic vs. biphasic); tumor size (≤ 2 cm vs. > 2 cm); and resectability (potentially resectable vs. unresectable). Multivariate logistic regression was performed to assess which of the following factors were associated with FN: clinical suspicion of PDAC; size >2 cm; presence of metastases; protocol; isoattenuating tumor; and potentially resectable disease on imaging. RESULTS In total, 176 CECTs (127 uniphasic; 49 biphasic) in 154 patients (90 men, mean age 72 ± 11 years) were included. Sensitivity was 125/149 (83.9%) overall and 87/106 (82.1%) and 38/43 (88.4%) for uniphasic and biphasic protocols, respectively. Sensitivity was decreased for tumors ≤ 2 cm (45.4% vs. 90.6%), no liver metastases (78.0% vs. 95.9%), and potentially resectable disease (65.3% vs. 93.0%). Factors significantly associated with FN were clinical suspicion (OR, 0.24, 95% CI: 0.07-0.75), size>2 cm (OR, 0.10, 95% CI: 0.02-0.44), absence of liver metastases (OR, 4.94, 95% CI: 1.29-22.99), and potentially resectable disease (OR, 4.13, 95% CI: 1.07-16.65). CONCLUSIONS In our population, the overall sensitivity of CECT to detect PDAC is 83.9%; however, this is substantially lower in several scenarios, including patients with potentially resectable disease. This finding has important implications for patient outcomes and efforts to maximize CECT sensitivity should be sought. CLINICAL RELEVANCE STATEMENT The sensitivity of CECT to detect PDAC is significantly decreased in the setting of sub-2 cm tumors and potentially resectable disease. A dedicated biphasic pancreatic CECT protocol has higher sensitivity and should be applied in patients with suspected pancreatic disease. KEY POINTS • The sensitivities of contrast-enhanced CT for the detection of PDAC were 87/106 (82.1%) and 38/43 (88.4%) for uniphasic and biphasic protocols, respectively. • The sensitivity of contrast-enhanced CT was decreased for small tumors ≤ 2 cm (45.4% vs. 90.6%), if there were no liver metastases (78.0% vs. 95.9%), and with potentially resectable disease (65.3% vs. 93.0%). • Absence of liver metastases (OR, 4.94, 95% CI: 1.29-22.99) and potentially resectable disease (OR, 4.13, 95% CI: 1.07-16.65) were associated with a false--negative (FN) CT result; suspicion of malignancy on the imaging requisition (OR, 0.24, 95% CI: 0.07-0.75) and size > 2 cm (OR, 0.10, 95% CI: 0.02-0.44) were negatively associated with FN.
Collapse
Affiliation(s)
- Max LeBlanc
- Department of Diagnostic Radiology, Queen Elizabeth II Health Sciences Centre and Dalhousie University, Victoria General Building, 3rd floor, 1276 South Park Street, Halifax, Nova Scotia, B3H 2Y9, Canada
| | - Jessie Kang
- Department of Diagnostic Radiology, Queen Elizabeth II Health Sciences Centre and Dalhousie University, Victoria General Building, 3rd floor, 1276 South Park Street, Halifax, Nova Scotia, B3H 2Y9, Canada
| | - Andreu F Costa
- Department of Diagnostic Radiology, Queen Elizabeth II Health Sciences Centre and Dalhousie University, Victoria General Building, 3rd floor, 1276 South Park Street, Halifax, Nova Scotia, B3H 2Y9, Canada.
| |
Collapse
|
11
|
Miyazawa M, Hirono S, Kawai M, Okada KI, Kitahata Y, Motobayashi H, Sato M, Yoshimura T, Ueno M, Hayami S, Miyamoto A, Shimizu A, Yamaue H. Radiographic duodenal invasion is associated with poor prognosis and early recurrence in patients with pancreatic ductal adenocarcinoma. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2023; 49:106960. [PMID: 37353425 DOI: 10.1016/j.ejso.2023.06.012] [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: 03/06/2023] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 06/25/2023]
Abstract
BACKGROUND The prognostic impact of radiographic duodenal invasion (rDI) of pancreatic ductal adenocarcinoma (PDAC) has yet to be fully elucidated. This retrospective study aimed to investigate the prognostic and clinicopathological significance of rDI in patients with PDAC after pancreatoduodenectomy (PD). MATERIALS AND METHODS We retrospectively analyzed 223 consecutive patients with resectable (R) and borderline resectable (BR)-PDAC that underwent up-front PD between 2002 and 2018. rDI was assessed by preoperative multi-detector row computed tomography. RESULTS Ninety-three (42%) patients with PDAC had rDI, and all of them had pathological DI (pDI). The rDI(+) group had larger tumor size, BR-PDAC was more common, there was higher serum CA19-9 level, and microscopic lymphovascular invasion was more common than in the rDI(-) group. rDI was associated with significant reduction in overall survival (OS) (P < 0.001) and recurrence-free survival (RFS) (P < 0.001). In multivariate analysis, rDI was an independent prognostic factor in OS [hazard ratio (HR) = 0.52; 95% confidence interval (CI) 0.38-0.73, P < 0.001] and RFS [HR = 0.56; 95% CI 0.40-0.78, P = 0.001]. rDI was also an independent risk factor for early recurrence within 12 months [odds ratio (OR) = 0.36; 95% CI 0.18-0.73, P = 0.005]. rDI had positive correlation with liver recurrence (P = 0.024). CONCLUSION Biological aggressiveness of PDAC with rDI implies short OS and early recurrence with frequent liver metastasis. Aggressive perioperative chemotherapy is recommended to improve prognosis, especially for R-PDAC patients with rDI.
Collapse
Affiliation(s)
- Motoki Miyazawa
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Seiko Hirono
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan; Division of Hepato-Biliary-Pancreatic Surgery, Department of Gastroenterological Surgery, Hyogo Medical University, Hyogo, Japan.
| | - Manabu Kawai
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Ken-Ichi Okada
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuji Kitahata
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Hideki Motobayashi
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Masatoshi Sato
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Tomohiro Yoshimura
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Masaki Ueno
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Shinya Hayami
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Atsushi Miyamoto
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Atsushi Shimizu
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan; Division of Hepato-Biliary-Pancreatic Surgery, Department of Gastroenterological Surgery, Hyogo Medical University, Hyogo, Japan
| | - Hiroki Yamaue
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
12
|
Ungkulpasvich U, Hatakeyama H, Hirotsu T, di Luccio E. Pancreatic Cancer and Detection Methods. Biomedicines 2023; 11:2557. [PMID: 37760999 PMCID: PMC10526344 DOI: 10.3390/biomedicines11092557] [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: 08/21/2023] [Revised: 09/05/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The pancreas is a vital organ with exocrine and endocrine functions. Pancreatitis is an inflammation of the pancreas caused by alcohol consumption and gallstones. This condition can heighten the risk of pancreatic cancer (PC), a challenging disease with a high mortality rate. Genetic and epigenetic factors contribute significantly to PC development, along with other risk factors. Early detection is crucial for improving PC outcomes. Diagnostic methods, including imagining modalities and tissue biopsy, aid in the detection and analysis of PC. In contrast, liquid biopsy (LB) shows promise in early tumor detection by assessing biomarkers in bodily fluids. Understanding the function of the pancreas, associated diseases, risk factors, and available diagnostic methods is essential for effective management and early PC detection. The current clinical examination of PC is challenging due to its asymptomatic early stages and limitations of highly precise diagnostics. Screening is recommended for high-risk populations and individuals with potential benign tumors. Among various PC screening methods, the N-NOSE plus pancreas test stands out with its high AUC of 0.865. Compared to other commercial products, the N-NOSE plus pancreas test offers a cost-effective solution for early detection. However, additional diagnostic tests are required for confirmation. Further research, validation, and the development of non-invasive screening methods and standardized scoring systems are crucial to enhance PC detection and improve patient outcomes. This review outlines the context of pancreatic cancer and the challenges for early detection.
Collapse
Affiliation(s)
| | | | | | - Eric di Luccio
- Hirotsu Bioscience Inc., 22F The New Otani Garden Court, 4-1 Kioi-cho, Chiyoda-ku, Tokyo 102-0094, Japan; (U.U.); (H.H.); (T.H.)
| |
Collapse
|
13
|
Adham S, Ferri M, Lee SY, Larocque N, Alwahbi OA, Ruo L, van der Pol CB. Pancreatic ductal adenocarcinoma (PDAC) regional nodal disease at standard lymphadenectomy: is MRI accurate for identifying node-positive patients? Eur Radiol 2023; 33:5976-5983. [PMID: 37004569 DOI: 10.1007/s00330-023-09597-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 04/04/2023]
Abstract
OBJECTIVE To determine the accuracy of qualitative and quantitative MRI features for the diagnosis of pathologic regional lymph nodes at standard lymphadenectomy in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS All adult patients with pancreatic MRI performed from 2011 to 2021 within 3 months of a pancreaticoduodenectomy were eligible for inclusion in this single-center retrospective cohort study. Regional nodes at standard lymphadenectomy were independently reviewed by two fellowship-trained abdominal radiologists for the following qualitative features: heterogeneous T2 signal, round shape, indistinct margin, peri-nodal fat stranding, and restricted diffusion greater than the spleen. Quantitative characteristics including primary tumor size, largest node short- and long-axes length, number of regional nodes, absolute apparent diffusion coefficient (ADC) values, and ADC node-to-spleen signal index were assessed. Analysis was at the patient-level with surgical pathology as the reference standard. RESULTS Of 75 patients, 85% (64/75) were positive for regional nodal disease on histopathology. None of the qualitative variables evaluated on MRI was associated with pathologic nodes. Median primary tumor maximum diameter was slightly larger for patients with pathologic nodes compared to those without (18 mm (10-42 mm) vs 16 mm (9-22 mm), p = 0.027). None of the other quantitative features was associated with pathologic nodes. Radiologist opinion was not associated with pathologic nodes (p = 0.520). Interobserver agreement was fair (kappa = 0.257). CONCLUSIONS Lymph node morphologic features and radiologist opinion using MRI are of limited value for diagnosing PDAC regional nodal disease. Improved diagnostic techniques are needed given the prognostic implications of pathologic lymph nodes in these patients. KEY POINTS • Multiple lymph node morphologic features routinely assessed on MRI for malignancies elsewhere in the body are likely not applicable when assessing for pancreatic ductal adenocarcinoma nodal disease. • Interobserver agreement for the presence or absence of pancreatic ductal adenocarcinoma lymph node morphologic features on MRI is fair (kappa = 0.257). • Many more lymph nodes are resected at PDAC standard lymphadenectomy than are detectable on MRI, median 25 vs 5 (p < 0.001), suggesting improved diagnostic techniques are needed to identify PDAC nodal disease.
Collapse
Affiliation(s)
- Sami Adham
- Department of Diagnostic Imaging, Juravinski Hospital and Cancer Centre, Hamilton Health Sciences, 711 Concession Street, Hamilton, ON, L8V 1C3, Canada
- Department of Radiology, McMaster University, Hamilton, ON, Canada
| | - Melanie Ferri
- Department of Diagnostic Imaging, Juravinski Hospital and Cancer Centre, Hamilton Health Sciences, 711 Concession Street, Hamilton, ON, L8V 1C3, Canada
- Department of Radiology, McMaster University, Hamilton, ON, Canada
| | - Stefanie Y Lee
- Department of Diagnostic Imaging, Juravinski Hospital and Cancer Centre, Hamilton Health Sciences, 711 Concession Street, Hamilton, ON, L8V 1C3, Canada
- Department of Radiology, McMaster University, Hamilton, ON, Canada
| | - Natasha Larocque
- Department of Radiology, McMaster University, Hamilton, ON, Canada
- Hamilton General Hospital, Hamilton Health Sciences, Hamilton, ON, Canada
| | - Omar A Alwahbi
- Department of Radiology, McMaster University, Hamilton, ON, Canada
| | - Leyo Ruo
- Department of Radiology, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Juravinski Hospital and Cancer Centre, Hamilton Health Sciences, Hamilton, ON, Canada
| | - Christian B van der Pol
- Department of Diagnostic Imaging, Juravinski Hospital and Cancer Centre, Hamilton Health Sciences, 711 Concession Street, Hamilton, ON, L8V 1C3, Canada.
- Department of Radiology, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
14
|
Abstract
Computed tomography (CT), MR imaging, and PET with fluorodeoxyglucose F18/CT are commonly used for radiation therapy planning; however, issues including precise nodal staging on CT or false positive results on PET/CT limit their usability. Clinical trials using fibroblast activation protein ligands for additional imaging have provided promising results regarding staging and target volume delineation-particularly suitable for sarcoma, some gastrointestinal tumors, head and neck tumors, and lung and pancreatic cancer. Although further prospective trials are necessary to identify clinical settings for its application in radiation oncology, fibroblast activation protein inhibitor PET/CT indisputably represents an excellent opportunity for assisting radiotherapy planning.
Collapse
Affiliation(s)
- Stefan A Koerber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center, Heidelberg, Germany; Department of Radiation Oncology, Barmherzige Brueder Hospital Regensburgh, Regensburg, Germany.
| |
Collapse
|
15
|
Jiang C, Yuan Y, Gu B, Ahn E, Kim J, Feng D, Huang Q, Song S. Preoperative prediction of microvascular invasion and perineural invasion in pancreatic ductal adenocarcinoma with 18F-FDG PET/CT radiomics analysis. Clin Radiol 2023:S0009-9260(23)00219-2. [PMID: 37365115 DOI: 10.1016/j.crad.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 04/23/2023] [Accepted: 05/13/2023] [Indexed: 06/28/2023]
Abstract
AIM To develop and validate a predictive model based on 2-[18F]-fluoro-2-deoxy-d-glucose (18F-FDG) positron-emission tomography (PET)/computed tomography (CT) radiomics features and clinicopathological parameters to preoperatively identify microvascular invasion (MVI) and perineural invasion (PNI), which are important predictors of poor prognosis in patients with pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS Preoperative 18F-FDG PET/CT images and clinicopathological parameters of 170 patients in PDAC were collected retrospectively. The whole tumour and its peritumoural variants (tumour dilated with 3, 5, and 10 mm pixels) were applied to add tumour periphery information. A feature-selection algorithm was employed to mine mono-modality and fused feature subsets, then conducted binary classification using gradient boosted decision trees. RESULTS For MVI prediction, the model performed best on a fused subset of 18F-FDG PET/CT radiomics features and two clinicopathological parameters, with an area under the receiver operating characteristic curve (AUC) of 83.08%, accuracy of 78.82%, recall of 75.08%, precision of 75.5%, and F1-score of 74.59%. For PNI prediction, the model achieved best prediction results only on the subset of PET/CT radiomics features, with AUC of 94%, accuracy of 89.33%, recall of 90%, precision of 87.81%, and F1 score of 88.35%. In both models, 3 mm dilation on the tumour volume produced the best results. CONCLUSIONS The radiomics predictors from preoperative 18F-FDG PET/CT imaging exhibited instructive predictive efficacy in the identification of MVI and PNI status preoperatively in PDAC. Peritumoural information was shown to assist in MVI and PNI predictions.
Collapse
Affiliation(s)
- C Jiang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China; Department of Nuclear Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Y Yuan
- Biomedical and Multimedia Information Technology Research Group, School of Computer Science, University of Sydney, Sydney, Australia
| | - B Gu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - E Ahn
- Discipline of Information Technology, College of Science & Engineering, James Cook University, Australia
| | - J Kim
- Biomedical and Multimedia Information Technology Research Group, School of Computer Science, University of Sydney, Sydney, Australia
| | - D Feng
- Biomedical and Multimedia Information Technology Research Group, School of Computer Science, University of Sydney, Sydney, Australia
| | - Q Huang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - S Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.
| |
Collapse
|
16
|
Fiore M, Coppola A, Petrianni GM, Trecca P, D’Ercole G, Cimini P, Ippolito E, Caputo D, Beomonte Zobel B, Coppola R, Ramella S. Advances in pre-treatment evaluation of pancreatic ductal adenocarcinoma: a narrative review. J Gastrointest Oncol 2023; 14:1114-1130. [PMID: 37201095 PMCID: PMC10186502 DOI: 10.21037/jgo-22-1034] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/08/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Despite advances in the multidisciplinary management of pancreatic cancer, overall prognosis remains poor, due to early progression of the disease. There is a need to also take action in staging, to make it increasingly accurate and complete, to define the setting of the therapeutic strategy. This review was planned to update the current status of pre-treatment evaluation for pancreatic cancer. METHODS We conducted an extensive review, including relevant articles dealing with traditional imaging, functional imaging and minimally invasive surgical procedures before treatment for pancreatic cancer. We searched articles written in English only. Data in the PubMed database, published in the period between January 2000 and January 2022, were retrieved. Prospective observational studies, retrospective analyses and meta-analyses were reviewed and analysed. KEY CONTENT AND FINDINGS Each imaging modality (endoscopic ultrasonography, endoscopic retrograde cholangiopancreatography, computed tomography, positron emission tomography/computed tomography, staging laparoscopy) has its own diagnostic advantages and limitations. The sensitivity, specificity and accuracy for each image set are reported. Data that support the increasing role of neoadjuvant therapy (radiotherapy and chemotherapy) and the meaning of a patient-tailored treatment selection, based on tumour staging, are also discussed. CONCLUSIONS A multimodal pre-treatment workup should be searched as it improves staging accuracy, orienting patients with resectable tumors towards surgery, optimizing patient selection with locally advanced tumors to neoadjuvant or definite therapy and avoiding surgical resection or curative radiotherapy in those with metastatic disease.
Collapse
Affiliation(s)
- Michele Fiore
- Research Unit of Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | | | - Gian Marco Petrianni
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Pasquale Trecca
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Gabriele D’Ercole
- Research Unit of Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Paola Cimini
- Operative Research Unit of Radiology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Edy Ippolito
- Research Unit of Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Damiano Caputo
- Department of Surgery and Research Unit of General Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Research Unit of General Surgery Unit Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Bruno Beomonte Zobel
- Operative Research Unit of Radiology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Roberto Coppola
- Department of Surgery and Research Unit of General Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Research Unit of General Surgery Unit Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Sara Ramella
- Research Unit of Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| |
Collapse
|
17
|
Liao H, Yang J, Li Y, Liang H, Ye J, Liu Y. One 3D VOI-based deep learning radiomics strategy, clinical model and radiologists for predicting lymph node metastases in pancreatic ductal adenocarcinoma based on multiphasic contrast-enhanced computer tomography. Front Oncol 2022; 12:990156. [PMID: 36158647 PMCID: PMC9500296 DOI: 10.3389/fonc.2022.990156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose We designed to construct one 3D VOI-based deep learning radiomics strategy for identifying lymph node metastases (LNM) in pancreatic ductal adenocarcinoma on the basis of multiphasic contrast-enhanced computer tomography and to assist clinical decision-making. Methods This retrospective research enrolled 139 PDAC patients undergoing pre-operative arterial phase and venous phase scanning examination between 2015 and 2021. A primary group (training group and validation group) and an independent test group were divided. The DLR strategy included three sections. (1) Residual network three dimensional-18 (Resnet 3D-18) architecture was constructed for deep learning feature extraction. (2) Least absolute shrinkage and selection operator model was used for feature selection. (3) Fully connected network served as the classifier. The DLR strategy was applied for constructing different 3D CNN models using 5-fold cross-validation. Radiomics scores (Rad score) were calculated for distinguishing the statistical difference between negative and positive lymph nodes. A clinical model was constructed by combining significantly different clinical variables using univariate and multivariable logistic regression. The manifestation of two radiologists was detected for comparing with computer-developed models. Receiver operating characteristic curves, the area under the curve, accuracy, precision, recall, and F1 score were used for evaluating model performance. Results A total of 45, 49, and 59 deep learning features were selected via LASSO model. No matter in which 3D CNN model, Rad score demonstrated the deep learning features were significantly different between non-LNM and LNM groups. The AP+VP DLR model yielded the best performance in predicting status of lymph node in PDAC with an AUC of 0.995 (95% CI:0.989-1.000) in training group; an AUC of 0.940 (95% CI:0.910-0.971) in validation group; and an AUC of 0.949 (95% CI:0.914-0.984) in test group. The clinical model enrolled the histological grade, CA19-9 level and CT-reported tumor size. The AP+VP DLR model outperformed AP DLR model, VP DLR model, clinical model, and two radiologists. Conclusions The AP+VP DLR model based on Resnet 3D-18 demonstrated excellent ability for identifying LNM in PDAC, which could act as a non-invasive and accurate guide for clinical therapeutic strategies. This 3D CNN model combined with 3D tumor segmentation technology is labor-saving, promising, and effective.
Collapse
Affiliation(s)
- Hongfan Liao
- College of Medical Informatics, Chongqing Medical University, Chongqing, China
| | - Junjun Yang
- Key Laboratory of Optoelectronic Technology and Systems of the Ministry of Education, Chongqing University, Chongqing, China
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongwei Liang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junyong Ye
- Key Laboratory of Optoelectronic Technology and Systems of the Ministry of Education, Chongqing University, Chongqing, China
| | - Yanbing Liu
- College of Medical Informatics, Chongqing Medical University, Chongqing, China
- *Correspondence: Yanbing Liu,
| |
Collapse
|
18
|
Radiolabeled FAP inhibitors as new pantumoral radiopharmaceuticals for PET imaging: a pictorial essay. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00506-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
19
|
Shi L, Wang L, Wu C, Wei Y, Zhang Y, Chen J. Preoperative Prediction of Lymph Node Metastasis of Pancreatic Ductal Adenocarcinoma Based on a Radiomics Nomogram of Dual-Parametric MRI Imaging. Front Oncol 2022; 12:927077. [PMID: 35875061 PMCID: PMC9298539 DOI: 10.3389/fonc.2022.927077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/06/2022] [Indexed: 12/12/2022] Open
Abstract
PurposeThis study aims to uncover and validate an MRI-based radiomics nomogram for detecting lymph node metastasis (LNM) in pancreatic ductal adenocarcinoma (PDAC) patients prior to surgery.Materials and MethodsWe retrospectively collected 141 patients with pathologically confirmed PDAC who underwent preoperative T2-weighted imaging (T2WI) and portal venous phase (PVP) contrast-enhanced T1-weighted imaging (T1WI) scans between January 2017 and December 2021. The patients were randomly divided into training (n = 98) and validation (n = 43) cohorts at a ratio of 7:3. For each sequence, 1037 radiomics features were extracted and analyzed. After applying the gradient-boosting decision tree (GBDT), the key MRI radiomics features were selected. Three radiomics scores (rad-score 1 for PVP, rad-score 2 for T2WI, and rad-score 3 for T2WI combined with PVP) were calculated. Rad-score 3 and clinical independent risk factors were combined to construct a nomogram for the prediction of LNM of PDAC by multivariable logistic regression analysis. The predictive performances of the rad-scores and the nomogram were assessed by the area under the operating characteristic curve (AUC), and the clinical utility of the radiomics nomogram was assessed by decision curve analysis (DCA).ResultsSix radiomics features of T2WI, eight radiomics features of PVP and ten radiomics features of T2WI combined with PVP were found to be associated with LNM. Multivariate logistic regression analysis showed that rad-score 3 and MRI-reported LN status were independent predictors. In the training and validation cohorts, the AUCs of rad-score 1, rad-score 2 and rad-score 3 were 0.769 and 0.751, 0.807 and 0.784, and 0.834 and 0.807, respectively. The predictive value of rad-score 3 was similar to that of rad-score 1 and rad-score 2 in both the training and validation cohorts (P > 0.05). The radiomics nomogram constructed by rad-score 3 and MRI-reported LN status showed encouraging clinical benefit, with an AUC of 0.845 for the training cohort and 0.816 for the validation cohort.ConclusionsThe radiomics nomogram derived from the rad-score based on MRI features and MRI-reported lymph status showed outstanding performance for the preoperative prediction of LNM of PDAC.
Collapse
Affiliation(s)
- Lin Shi
- Cancer Center, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Ling Wang
- Cancer Center, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Cuiyun Wu
- Cancer Center, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Yuguo Wei
- Precision Health Institution, General Electric Healthcare, Hangzhou, China
| | - Yang Zhang
- Cancer Center, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Junfa Chen
- Cancer Center, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- *Correspondence: Junfa Chen,
| |
Collapse
|
20
|
Gnanasegaran G, Agrawal K, Wan S. 18F-Fluorodeoxyglucose-PET-Computerized Tomography and non-Fluorodeoxyglucose PET-Computerized Tomography in Hepatobiliary and Pancreatic Malignancies. PET Clin 2022; 17:369-388. [PMID: 35717098 DOI: 10.1016/j.cpet.2022.03.007] [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: 02/07/2023]
Abstract
Molecular imaging with PET-computerized tomography (PET-CT) plays an important role in oncology. There is current and evolving evidence supporting the use of fluorodeoxyglucose (FDG) and non-FDG tracers in assessment patients with hepatobiliary and pancreatic cancers in various clinical scenarios. In this chapter, we discuss the advantages and limitations of FDG and non-FDG PET-CT in the management of patients with hepatobiliary and pancreatic cancers.
Collapse
Affiliation(s)
| | | | - Simon Wan
- University College London Hospitals NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
21
|
Zhang Z, Jia G, Pan G, Cao K, Yang Q, Meng H, Yang J, Zhang L, Wang T, Cheng C, Zuo C. Comparison of the diagnostic efficacy of 68 Ga-FAPI-04 PET/MR and 18F-FDG PET/CT in patients with pancreatic cancer. Eur J Nucl Med Mol Imaging 2022; 49:2877-2888. [PMID: 35243518 DOI: 10.1007/s00259-022-05729-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 02/13/2022] [Indexed: 12/26/2022]
Abstract
PURPOSE We sought to assess the performance of 68 Ga-FAPI-04 PET/MR for the diagnosis of primary tumours as well as metastatic lesions in patients with pancreatic cancer and to compare the results with those of 18F-FDG PET/CT. METHODS Prospectively, we evaluated 33 patients suspected to have pancreatic adenocarcinoma, of whom thirty-two were confirmed by histopathology, and one had autoimmune pancreatitis confirmed by needle biopsy and glucocorticoid treatment. Within 1 week, each patient underwent both 68 Ga-FAPI-04 PET/MR and 18F-FDG PET/CT. Comparisons of the detection abilities for primary tumours, lymph nodes, and metastases were conducted for the two imaging approaches. The original maximum standard uptake values (SUVmax) and normalised SUVmax (SUVmax/SUVbkgd) of paired lesions on 68 Ga-FAPI-04 PET/MR and 18F-FDG PET/CT were measured and compared. RESULTS Thirty pancreatic cancer patients and three pancreatitis patients were enrolled. 68 Ga-FAPI-04 PET/MR and 18F-FDG PET/CT exhibited equivalent (100%) detection rates for primary tumours. The original/normalised SUVmax of primary tumours on 68 Ga-FAPI-04 PET was markedly higher than that on 18F-FDG (p < 0.05). Sixteen pancreatic cancer patients had pancreatic parenchymal uptake, whereas 18F-FDG PET images showed parenchymal uptake in only four patients (53.33% vs. 13.33%, p < 0.001). 68 Ga-FAPI-04 PET detected more positive lymph nodes than 18F-FDG PET (42 vs. 30, p < 0.001), while 18F-FDG PET was able to detect more liver metastases than 68 Ga-FAPI-04 (181 vs. 104, p < 0.001). In addition, multisequence MR imaging helped explain ten pancreatic cancers that could not be definitively revealed due to 68 Ga-FAPI-04 inflammatory uptake and identified more liver metastases than 18F-FDG (256 vs. 181, p < 0.001). CONCLUSION 68 Ga-FAPI-04 PET might be better than 18F-FDG PET in the detection of suspicious lymph node metastases. MR multiple sequence imaging of 68 Ga-FAPI-04 PET/MR was helpful for explaining pancreatic lesions in patients with obstructive inflammation and detecting tiny liver metastases.
Collapse
Affiliation(s)
- Zeyu Zhang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Guorong Jia
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Guixia Pan
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Kai Cao
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Qinqin Yang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Hongyu Meng
- Department of Radiology, Shanghai Fourth People's Hospital Affiliated to Tongji University, School of Medicine, Shanghai, 200434, China
| | - Jian Yang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Lu Zhang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Tao Wang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Chao Cheng
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Changjing Zuo
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
22
|
Utility of 18F-FDG PET/CT in Management of Pancreatic and Periampullary Masses—Prospective Study from a Tertiary Care Center. Indian J Surg Oncol 2022; 13:288-298. [DOI: 10.1007/s13193-021-01434-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/05/2021] [Indexed: 12/24/2022] Open
|
23
|
Pande R, Chughtai S, Ahuja M, Brown R, Bartlett DC, Dasari BV, Marudanayagam R, Mirza D, Roberts K, Isaac J, Sutcliffe RP, Chatzizacharias NA. Para-aortic lymph node involvement should not be a contraindication to resection of pancreatic ductal adenocarcinoma. World J Gastrointest Surg 2022; 14:429-441. [PMID: 35734625 PMCID: PMC9160687 DOI: 10.4240/wjgs.v14.i5.429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/19/2022] [Accepted: 04/21/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Para-aortic lymph nodes (PALN) are found in the aortocaval groove and they are staged as metastatic disease if involved by pancreatic ductal adenocarcinoma (PDAC). The data in the literature is conflicting with some studies having associated PALN involvement with poor prognosis, while others not sharing the same results. PALN resection is not included in the standard lymphadenectomy during pancreatic resections as per the International Study Group for Pancreatic Surgery and there is no consensus on the management of these cases. AIM To investigate the prognostic significance of PALN metastases on the oncological outcomes after resection for PDAC. METHODS This is a retrospective cohort study of data retrieved from a prospectively maintained database on consecutive patients undergoing pancreatectomies for PDAC where PALN was sampled between 2011 and 2020. Statistical comparison of the data between PALN+ and PALN- subgroups, survival analysis with the Kaplan-Meier method and risk analysis with univariable and multivariable time to event Cox regression analysis were performed, specifically assessing oncological outcomes such as median overall survival (OS) and disease-free survival (DFS). RESULTS 81 cases had PALN sampling and 17 (21%) were positive. Pathological N stage was significantly different between PALN+ and PALN- patients (P = 0.005), while no difference was observed in any of the other characteristics. Preoperative imaging diagnosed PALN positivity in one case. OS and DFS were comparable between PALN+ and PALN- patients with lymph node positive disease (OS: 13.2 mo vs 18.8 mo, P = 0.161; DFS: 13 mo vs 16.4 mo, P = 0.179). No difference in OS or DFS was identified between PALN positive and negative patients when they received chemotherapy either in the neoadjuvant or in the adjuvant setting (OS: 23.4 mo vs 20.6 mo, P = 0.192; DFS: 23.9 mo vs 20.5 mo, P = 0.718). On the contrary, when patients did not receive chemotherapy, PALN disease had substantially shorter OS (5.5 mo vs 14.2 mo; P = 0.015) and DFS (4.4 mo vs 9.8 mo; P < 0.001). PALN involvement was not identified as an independent predictor for OS after multivariable analysis, while it was for DFS doubling the risk of recurrence. CONCLUSION PALN involvement does not affect OS when patients complete the indicated treatment pathway for PDAC, surgery and chemotherapy, and should not be considered as a contraindication to resection.
Collapse
Affiliation(s)
- Rupaly Pande
- Department of Hepato-Pancreato-Biliary and Liver Transplant Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, United Kingdom
| | - Shafiq Chughtai
- Department of Hepato-Pancreato-Biliary and Liver Transplant Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, United Kingdom
| | - Manish Ahuja
- Department of Hepato-Pancreato-Biliary and Liver Transplant Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, United Kingdom
| | - Rachel Brown
- Department of Pathology, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, United Kingdom
| | - David C Bartlett
- Department of Hepato-Pancreato-Biliary and Liver Transplant Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, United Kingdom
| | - Bobby V Dasari
- Department of Hepato-Pancreato-Biliary and Liver Transplant Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, United Kingdom
| | - Ravi Marudanayagam
- Department of Hepato-Pancreato-Biliary and Liver Transplant Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, United Kingdom
| | - Darius Mirza
- Department of Hepato-Pancreato-Biliary and Liver Transplant Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, United Kingdom
| | - Keith Roberts
- Department of Hepato-Pancreato-Biliary and Liver Transplant Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, United Kingdom
| | - John Isaac
- Department of Hepato-Pancreato-Biliary and Liver Transplant Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, United Kingdom
| | - Robert P Sutcliffe
- Department of Hepato-Pancreato-Biliary and Liver Transplant Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, United Kingdom
| | - Nikolaos A Chatzizacharias
- Department of Hepato-Pancreato-Biliary and Liver Transplant Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, United Kingdom
| |
Collapse
|
24
|
Zakaria A, Al-Share B, Klapman JB, Dam A. The Role of Endoscopic Ultrasonography in the Diagnosis and Staging of Pancreatic Cancer. Cancers (Basel) 2022; 14:1373. [PMID: 35326524 PMCID: PMC8946253 DOI: 10.3390/cancers14061373] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is the fourth leading cause of cancer-related death and the second gastrointestinal cancer-related death in the United States. Early detection and accurate diagnosis and staging of pancreatic cancer are paramount in guiding treatment plans, as surgical resection can provide the only potential cure for this disease. The overall prognosis of pancreatic cancer is poor even in patients with resectable disease. The 5-year survival after surgical resection is ~10% in node-positive disease compared to ~30% in node-negative disease. The advancement of imaging studies and the multidisciplinary approach involving radiologists, gastroenterologists, advanced endoscopists, medical, radiation, and surgical oncologists have a major impact on the management of pancreatic cancer. Endoscopic ultrasonography is essential in the diagnosis by obtaining tissue (FNA or FNB) and in the loco-regional staging of the disease. The advancement in EUS techniques has made this modality a critical adjunct in the management process of pancreatic cancer. In this review article, we provide an overall description of the role of endoscopic ultrasonography in the diagnosis and staging of pancreatic cancer.
Collapse
Affiliation(s)
- Ali Zakaria
- Department of Gastroenterology-Advanced Endoscopy, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA; (J.B.K.); (A.D.)
| | - Bayan Al-Share
- Department of Hematology and Oncology, Karmanos Cancer Center, Wayne State University, Detroit, MI 48201, USA;
| | - Jason B. Klapman
- Department of Gastroenterology-Advanced Endoscopy, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA; (J.B.K.); (A.D.)
| | - Aamir Dam
- Department of Gastroenterology-Advanced Endoscopy, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA; (J.B.K.); (A.D.)
| |
Collapse
|
25
|
Peng D, He J, Liu H, Cao J, Wang Y, Chen Y. FAPI PET/CT research progress in digestive system tumours. Dig Liver Dis 2022; 54:164-169. [PMID: 34364808 DOI: 10.1016/j.dld.2021.07.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 12/24/2022]
Abstract
18F-fluorodeoxyglucose positron emission tomography/computed tomography has been used in clinical practice for many years. This modality is of great value for tumour diagnosis, staging, and efficacy evaluations, but it has many limitations in the diagnosis and treatment of digestive system tumours. Fibroblast activation protein is highly expressed in gastrointestinal tumours. Various isotope-labelled fibroblast activation protein inhibitors are widely used in clinical research. These inhibitors have low background uptake in the brain, liver and oral/pharyngeal mucosa and show good contrast between the tumour and background, which makes up for the lack of fluorodeoxyglucose in the diagnosis of digestive system tumours. It better displays the primary tumours, metastases and regional lymph nodes of digestive system tumours, such as oesophageal cancer, gastric cancer and liver cancer, and also provides a new method for treating these tumours. Based on this background, this article introduces the current research status of fibroblast activation protein inhibitor positron emission tomography/computed tomography in various types of digestive system malignant tumours to provide more valuable information for diagnosing and treating digestive system tumours.
Collapse
Affiliation(s)
- Dengsai Peng
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No 25 TaiPing St, Jiangyang District, Luzhou, Sichuan 646000, PR China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, PR China; Academician (Expert) Workstation of Sichuan Province, 646000, PR China
| | - Jing He
- Department of Ultrasonography, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Hanxiang Liu
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No 25 TaiPing St, Jiangyang District, Luzhou, Sichuan 646000, PR China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, PR China; Academician (Expert) Workstation of Sichuan Province, 646000, PR China
| | - Jianpeng Cao
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No 25 TaiPing St, Jiangyang District, Luzhou, Sichuan 646000, PR China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, PR China; Academician (Expert) Workstation of Sichuan Province, 646000, PR China
| | - Yingwei Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No 25 TaiPing St, Jiangyang District, Luzhou, Sichuan 646000, PR China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, PR China; Academician (Expert) Workstation of Sichuan Province, 646000, PR China
| | - Yue Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No 25 TaiPing St, Jiangyang District, Luzhou, Sichuan 646000, PR China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, PR China; Academician (Expert) Workstation of Sichuan Province, 646000, PR China.
| |
Collapse
|
26
|
Yang T, Ma L, Hou H, Gao F, Tao W. FAPI PET/CT in the Diagnosis of Abdominal and Pelvic Tumors. Front Oncol 2022; 11:797960. [PMID: 35059319 PMCID: PMC8763785 DOI: 10.3389/fonc.2021.797960] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/23/2021] [Indexed: 12/11/2022] Open
Abstract
Positron emission tomography/computed tomography (PET/CT) with 18F-fluorodeoxyglucose (18F-FDG) is currently a standard imaging examination used in clinical practice, and plays an essential role in preoperative systemic evaluation and tumor staging in patients with tumors. However, 18F-FDG PET/CT has certain limitations in imaging of some tumors, like gastric mucus adenocarcinoma, highly differentiated hepatocellular carcinoma, renal cell carcinoma, and peritoneal metastasis. Therefore, to search for new tumor diagnosis methods has always been an important topic in radiographic imaging research. Fibroblast activation protein (FAP) is highly expressed in many epithelial carcinomas, and various isotope-labelled fibroblast activation protein inhibitors (FAPI) show lower uptake in the brain and abdominal tissues than in tumor, thus achieving high image contrast and good tumor delineation. In addition to primary tumors, FAPI PET/CT is better than FDG PET/CT for detecting lymph nodes and metastases. Additionally, the highly selective tumor uptake of FAPI may open up new application areas for the non-invasive characterization, staging of tumors, as well as monitoring tumor treatment efficacy. This review focuses on the recent research progress of FAPI PET/CT in the application to abdominal and pelvic tumors, with the aim of providing new insights for diagnostic strategies for tumor patients, especially those with metastases.
Collapse
Affiliation(s)
- Tianshuo Yang
- Department of Nuclear Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Long Ma
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Haodong Hou
- Key Laboratory for Experimental Teratology of the Ministry of Education and Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Feng Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Weijing Tao
- Department of Nuclear Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| |
Collapse
|
27
|
PET imaging of pancreatic cancer. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00207-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
28
|
Tabacchi E, Nanni C, Bossert I, Maffione AM, Fanti S. Diagnostic Applications of Nuclear Medicine: Pancreatic Cancer. NUCLEAR ONCOLOGY 2022:891-917. [DOI: 10.1007/978-3-031-05494-5_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
29
|
Gao H, Wang W, Yu X, Liu L. Patterns and predictors of pancreatic neuroendocrine tumor prognosis: Are no two leaves alike? Crit Rev Oncol Hematol 2021; 167:103493. [PMID: 34653597 DOI: 10.1016/j.critrevonc.2021.103493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 02/20/2021] [Accepted: 09/08/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic neuroendocrine tumors (PanNETs) are heterogeneous; thus, individual prognostic prediction is important. Clinicopathological features, like TNM stage, grade, and differentiation, are independent clinical predictors. However, single predictors are insufficient, as patients sharing similar clinicopathological features usually show distinct prognoses. Accordingly, novel nomograms and risk stratifications have been developed for more accurate PanNET prognostic prediction. Moreover, the exploration of molecular mechanisms has identified novel prognostic predictors for PanNET. Multi-analyte assays of molecular biomarkers provide a deeper understanding of PanNET features; however, the priority, and the optimal combination of classic and novel predictors for PanNET prognosis prediction remain unclear. In this review, we summarized the patterns and predictors of PanNET prognosis and discussed their clinical utility; we emphasized that PanNET at different stages have different superior predictor, and that multi-analyte assays are more sensitive than mono-analyte biomarkers. Therefore, combined biomarkers improve the accuracy of surveillance and optimize decision-making in clinical practice.
Collapse
Affiliation(s)
- HeLi Gao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 20032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China
| | - WenQuan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 20032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China
| | - XianJun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 20032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China.
| | - Liang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 20032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China.
| |
Collapse
|
30
|
Huang S, Chong H, Sun X, Wu Z, Jia Q, Zhang Y, Lan X. The Value of 18F-FDG PET/CT in Diagnosing Pancreatic Lesions: Comparison With CA19-9, Enhanced CT or Enhanced MR. Front Med (Lausanne) 2021; 8:668697. [PMID: 34692714 PMCID: PMC8531126 DOI: 10.3389/fmed.2021.668697] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 09/13/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: To investigate the value of 18F-FDG PET/CT in diagnosing pancreatic lesions, and compare it with CA19-9, contrast-enhanced CT (CECT), and contrast-enhanced MRI (CEMR). Methods: Cases of patients with suspected pancreatic lesions examined between January 1, 2011 and June 30, 2017 were retrospectively analyzed. CA19-9, CECT and CEMR within 2 weeks of PET/CT were evaluated. We compared the diagnostic efficacy of PET/CT with CA19-9, CECT and CEMR as well as combined tests. Results: A total of 467 cases were examined in this study, including 293 males and 174 females, with an average age of 57.79 ± 12.68 y (16-95 y). Cases in the malignant group (n = 248) had significantly higher SUVmax (7.34 ± 4.17 vs. 1.70 ± 2.68, P < 0.001) and CA19-9 (663.21 ± 531.98 vs. 87.80 ± 218.47, P < 0.001) than those in the benign group (n = 219). The sensitivity, specificity and accuracy of PET/CT were 91.9, 96.3, and 94.0%, respectively. Those for CECT were 83.6, 77.8, 81.2%, respectively; and 91.2, 75.0, 81.7% were for CEMR. PET/CT corrected 14.7% (28/191) CECT diagnoses and 12.2% (10/82) CEMR diagnoses. Although the diagnostic efficiency of CA19-9 was acceptable (80.0, 69.0, 74.9% respectively), the joint application of PET/CT and CA19-9 could significantly enhance the diagnostic efficiency compared with PET/CT alone (sen 97.4 vs. 90.5%, P = 0.0003; spe 100.0 vs. 95.2%, P = 0.0047). Conclusions: PET/CT has sensitivity similar to CECT, CEMR and significantly higher specificity and accuracy, helping reduce false diagnoses of morphological images. Combining PET/CT with CA19-9 could enhance diagnostic efficiency.
Collapse
Affiliation(s)
- Shengyun Huang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Huanhuan Chong
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, China.,Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xun Sun
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Zhijian Wu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Qing Jia
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
31
|
Korean clinical practice guideline for pancreatic cancer 2021: A summary of evidence-based, multi-disciplinary diagnostic and therapeutic approaches. Pancreatology 2021; 21:1326-1341. [PMID: 34148794 DOI: 10.1016/j.pan.2021.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/20/2021] [Accepted: 05/04/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer is the eighth most common cancer and the fifth most common cause of cancer-related death in Korea. To enable standardization of management and facilitate improvements in outcome, a total of 53 multi-disciplinary experts in gastroenterology, surgery, medical oncology, radiation oncology, radiology, nuclear medicine, and pathology in Korea developed new recommendations that integrate the most up-to-date, evidence-based research findings and expert opinions. Recommendations were made on imaging diagnosis, endoscopic management, surgery, radiotherapy, palliative chemotherapy, and specific management procedures, including neoadjuvant treatment or adjuvant treatment for patients with resectable, borderline resectable, and locally advanced unresectable pancreatic cancer. This is the English version of the Korean clinical practice guideline for pancreatic cancer 2021. This guideline includes 20 clinical questions and 32 statements. This guideline represents the most standard guideline for the diagnosis and treatment of patients with pancreatic ductal adenocarcinoma in adults at this time in Korea. The authors believe that this guideline will provide useful and informative advice.
Collapse
|
32
|
Ghidini M, Vuozzo M, Galassi B, Mapelli P, Ceccarossi V, Caccamo L, Picchio M, Dondossola D. The Role of Positron Emission Tomography/Computed Tomography (PET/CT) for Staging and Disease Response Assessment in Localized and Locally Advanced Pancreatic Cancer. Cancers (Basel) 2021; 13:4155. [PMID: 34439307 PMCID: PMC8394552 DOI: 10.3390/cancers13164155] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/03/2021] [Accepted: 08/16/2021] [Indexed: 11/25/2022] Open
Abstract
Pancreatic Cancer (PC) has a poor prognosis, with a 5-year survival rate of only 9%. Even after radical surgical procedures, PC patients have poor survival rates, with a high chance of relapse (70-80%). Imaging is involved in all aspects of the clinical management of PC, including detection and characterization of primary tumors and their resectability, assessment of vascular, perineural and lymphatic invasion and detection of distant metastases. The role of Positron Emission Tomography/Computed Tomography (PET/CT) in detecting PC is still controversial, with the international guidelines not recommending its routine use. However, in resectable PC, PET/CT may play a role in assessing PC stage and grade and potential resectability after neoadjuvant treatment. Quantitative image analysis (radiomics) and new PET/CT radiotracers account for future developments in metabolic imaging and may further improve the relevance of this technique in several aspects of PC. In the present review, the current state of the art and future directions of PET/CT in resectable PC are presented.
Collapse
Affiliation(s)
- Michele Ghidini
- Operative Unit of Oncology, Internal Medicine, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Marta Vuozzo
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany;
- University Medical Center, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Barbara Galassi
- Operative Unit of Oncology, Internal Medicine, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Paola Mapelli
- Vita-Salute San Raffaele University, 20132 Milan, Italy; (P.M.); (M.P.)
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Virginia Ceccarossi
- Dipartimento di Chirurgia Generale e dei Trapianti di Fegato, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (V.C.); (L.C.); (D.D.)
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, 20122 Milan, Italy
| | - Lucio Caccamo
- Dipartimento di Chirurgia Generale e dei Trapianti di Fegato, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (V.C.); (L.C.); (D.D.)
| | - Maria Picchio
- Vita-Salute San Raffaele University, 20132 Milan, Italy; (P.M.); (M.P.)
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Daniele Dondossola
- Dipartimento di Chirurgia Generale e dei Trapianti di Fegato, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (V.C.); (L.C.); (D.D.)
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, 20122 Milan, Italy
| |
Collapse
|
33
|
Gupta N, Yelamanchi R. Pancreatic adenocarcinoma: A review of recent paradigms and advances in epidemiology, clinical diagnosis and management. World J Gastroenterol 2021; 27:3158-3181. [PMID: 34163104 PMCID: PMC8218366 DOI: 10.3748/wjg.v27.i23.3158] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/03/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the dreaded malignancies for both the patient and the clinician. The five-year survival rate of pancreatic adenocarcinoma (PDA) is as low as 2% despite multimodality treatment even in the best hands. As per the Global Cancer Observatory of the International Agency for Research in Cancer estimates of pancreatic cancer, by 2040, a 61.7% increase is expected in the total number of cases globally. With the widespread availability of next-generation sequencing, the entire genome of the tumors is being sequenced regularly, providing insight into their pathogenesis. As invasive PDA arises from pancreatic intraepithelial neoplasia and mucinous neoplasm and intraductal papillary neoplasm, screening for them can be beneficial as the disease is curable with resection at an early stage. Routine preoperative biliary drainage has no role in patients suffering from PDA with obstructive jaundice. If performed, metallic stents are preferred over plastic ones. Minimally invasive procedures are preferred to open procedures as they have less morbidity. The duct-to-mucosa technique for pancreaticojejunostomy is presently widely practiced. The role of intraperitoneal drains after surgery for PDA is controversial. Neoadjuvant chemoradiotherapy has been proven to have a significant role both in locally advanced as well as in resectable PDA. Many new regimens and drugs have been added in the arsenal of chemoradiotherapy for metastatic disease. The roles of immunotherapy and gene therapy in PDA are being investigated. This review article is intended to improve the understanding of the readers with respect to the latest updates of PDA, which may help to trigger new research ideas and make better management decisions.
Collapse
Affiliation(s)
- Nikhil Gupta
- Department of Surgery, Atal Bihari Vajpayee Institute of Medical Sciences and Dr. Ram Manohar Lohia Hospital, Delhi 110001, India
| | - Raghav Yelamanchi
- Department of Surgery, Atal Bihari Vajpayee Institute of Medical Sciences and Dr. Ram Manohar Lohia Hospital, Delhi 110001, India
| |
Collapse
|
34
|
Zeeshan MS, Ramzan Z. Current controversies and advances in the management of pancreatic adenocarcinoma. World J Gastrointest Oncol 2021; 13:472-494. [PMID: 34163568 PMCID: PMC8204360 DOI: 10.4251/wjgo.v13.i6.472] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/22/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic adenocarcinoma is a lethal disease with a mortality rate that has not significantly improved over decades. This is likely due to several challenges unique to pancreatic cancer. Most patients with pancreatic cancer are diagnosed at a late stage of disease due to the lack of specific symptoms prompting an early investigation. A small subset of patients who are diagnosed at an early stage have a better chance at survival with curative surgical resection, but most patients still succumb to the disease in a few years. The dismal overall prognosis is due to suspected micro-metastasis at an early stage. Due to this reason, there is a recent interest in treating all patients with pancreatic cancers with systemic therapy upfront (including the ones that are surgically resectable). This approach is still not the standard of care due to the lack of robust prospective data available. Recent advancements in treatment regimens of chemotherapy, radiation and immunotherapy have improved the overall short-term survival but the long-term survival still remains poor. Novel approaches in diagnosis and treatment have shown promise in clinical studies but long-term clinical data is lacking. The following manuscript presents an overview of the epidemiology, diagnosis, staging, recent advances, novel approaches and controversies in the management of pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Muhammad Shehroz Zeeshan
- Gastrointestinal Section, Department of Medicine, Texas Health Harris Methodist Hospital, Fort Worth, TX 76104, United States
| | - Zeeshan Ramzan
- Gastrointestinal Section, Department of Medicine, Texas Health Harris Methodist Hospital, Fort Worth, TX 76104, United States
| |
Collapse
|
35
|
Xia J, Zhang H, Guan Q, Wang S, Li Y, Xie J, Li M, Huang H, Yan H, Chen T. Qualitative diagnostic signature for pancreatic ductal adenocarcinoma based on the within-sample relative expression orderings. J Gastroenterol Hepatol 2021; 36:1714-1720. [PMID: 33150986 DOI: 10.1111/jgh.15326] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/18/2020] [Accepted: 10/24/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) accounts for about 90% of pancreatic cancer, which is one of the most aggressive malignant neoplasms with a 9.3% five-year survival rate. The pathological biopsy is the current golden standard for confirming suspicious lesions of PDAC, but it is not entirely reliable because of the insufficient sampling amount and inaccurate sampling location. Therefore, developing a robust signature to aid the accurate diagnosis of PDAC is critical. METHODS Based on the within-sample relative expression orderings of gene pairs, we identified a qualitative signature to discriminate both PDAC and adjacent samples from both chronic pancreatitis and normal samples in the training datasets and validated it in other independent datasets produced by different laboratories with different measuring platforms. RESULTS A six-gene-pair signature was identified in the training data and validated in eight independent datasets. For surgical samples, 96.63% of 356 PDAC tissues, 100% of 11 pancreatitis tissues of non-cancer patients, and 23 of 24 normal pancreatic tissues were correctly classified. Especially, 59 of 60 cancer-adjacent normal tissues of PDAC patients were correctly identified as PDAC. For biopsy samples, all of 11 PDAC biopsy tissues were correctly classified as PDAC. CONCLUSION The signature can distinguish both PDAC and PDAC-adjacent normal tissues from both chronic pancreatitis and normal tissues of non-cancer patients even when the sampling locations are inaccurate, which can aid the diagnosis of PDAC.
Collapse
Affiliation(s)
- Jie Xia
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Huarong Zhang
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Qingzhou Guan
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Shanshan Wang
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yawei Li
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Jiajing Xie
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Meifeng Li
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Haiyan Huang
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Haidan Yan
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Ting Chen
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
36
|
Kumar R, Sharma A, Garg P, Sharma R, Datta-Gupta S. Role of dual-point 18F-FDG-PET/CT in the diagnosis of pancreatic adenocarcinoma, in patients with and without concomitant chronic pancreatitis: Comparison with CECT and EUS. Pancreatology 2021; 21:746-754. [PMID: 33632664 DOI: 10.1016/j.pan.2021.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/31/2020] [Accepted: 02/11/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE The aim of this study was to evaluate the diagnostic performance of dual-time-point-PET/CT, CECT and EUS + FNA in diagnosing pancreatic-ductal-adenocarcinomas (PDAC), in context of concomitant Chronic Pancreatitis (CP). METHODS 18F-FDG-PET/CTs were prospectively acquired in 22 confirmed CP and 23 confirmed PDAC patients (calculated for 90% power); and cut-offs of 2.2 for early-SUV(∼1hr), 2.4 for delayed-SUV(∼3hr) and 1.36 for Retention-index (RI), were derived. These cut-offs were validated in PET/CTs of 75 patients (51.9 ± 13.3years; 54 men) with pancreatic masses of unknown nature. Comparisons were made with triple-phase-CECT (73 patients) and EUS + FNA (54 patients). Histopathology was obtained in 68 patients (including all PDACs) and 7 were followed up for minimum of 2 years. RESULTS In patients without concomitant CP, sensitivity, specificity and accuracy for diagnosing malignancy in standard-acquisition-PET/CT, dual-time-point-PET/CT, CECT and EUS + FNA were 97.4%, 83.3%, 94.0%; 97.4%, 75.0%, 92%; 94.6%, 66.7%, 87.8% and 92.6%, 88.9%, 91.7% respectively. Corresponding values in patients with concomitant CP were 88.9%, 57.1%, 80.0%; 100%, 57.1%, 88%; 82.4%, 57.1%, 75% and 100%, 100%, 100% respectively. In lesions ≤2 cm (AJCC-T1), dual-time-point-PET/CT was the most sensitive (95.8%). ROC-analysis revealed significantly higher area-under-the-curve for RI over early-SUV (p = 0.002) in cases with concomitant CP only. In patients with confirmed liver-metastases, PET/CT and CECT identified 15/16 and 13/16 lesions. PET/CT identified additional lung-metastases in 3 and bone-metastasis in one patient. CONCLUSION In patients without concomitant CP and with larger lesions, PET/CT and CECT are equivocal as screening modalities, with no benefit of dual-time-point-PET/CT acquisitions. However, in patients with concomitant CP and smaller lesions, dual-time-point PET/CT is better; with sensitivity comparable to EUS + FNA.
Collapse
Affiliation(s)
- Rakesh Kumar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India.
| | - Anshul Sharma
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Pramod Garg
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - Raju Sharma
- Department of Radio-diagnosis, All India Institute of Medical Sciences, New Delhi, India
| | - S Datta-Gupta
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
37
|
Kato T, Hiraishi Y, Kaseda K, Suzuki S, Tanaka G, Yamauchi Y, Yasui M, Shinozaki-Ushiku A, Ushiku T, Yamaji O, O'Neal WK, Nagase T. A 79-Year-Old Man With Progressive Dyspnea and Multiple Pulmonary Nodules. Chest 2021; 158:e79-e84. [PMID: 32768080 DOI: 10.1016/j.chest.2020.03.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/08/2020] [Accepted: 03/29/2020] [Indexed: 11/29/2022] Open
Affiliation(s)
- Takafumi Kato
- Departments of Respiratory Medicine, The University of Tokyo Hospital, Tokyo, Japan; Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Yoshihisa Hiraishi
- Departments of Respiratory Medicine, The University of Tokyo Hospital, Tokyo, Japan.
| | - Ken Kaseda
- Departments of Respiratory Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Satoshi Suzuki
- Departments of Respiratory Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Goh Tanaka
- Departments of Respiratory Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Yasuhiro Yamauchi
- Departments of Respiratory Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Mariko Yasui
- Departments of Pathology, The University of Tokyo Hospital, Tokyo, Japan; Department of Pathology, School of Medicine, Teikyo University, Tokyo, Japan
| | | | - Tetsuo Ushiku
- Departments of Pathology, The University of Tokyo Hospital, Tokyo, Japan
| | - Osamu Yamaji
- Department of Internal Medicine, Toshima Hospital of Tokyo Metropolitan Health and Hospitals Corporation, Tokyo, Japan
| | - Wanda K O'Neal
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Takahide Nagase
- Departments of Respiratory Medicine, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
38
|
Gallo M, Adinolfi V, Morviducci L, Acquati S, Tuveri E, Ferrari P, Zatelli MC, Faggiano A, Argentiero A, Natalicchio A, D'Oronzo S, Danesi R, Gori S, Russo A, Montagnani M, Beretta GD, Di Bartolo P, Silvestris N, Giorgino F. Early prediction of pancreatic cancer from new-onset diabetes: an Associazione Italiana Oncologia Medica (AIOM)/Associazione Medici Diabetologi (AMD)/Società Italiana Endocrinologia (SIE)/Società Italiana Farmacologia (SIF) multidisciplinary consensus position paper. ESMO Open 2021; 6:100155. [PMID: 34020401 PMCID: PMC8144346 DOI: 10.1016/j.esmoop.2021.100155] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer (PC) is a common cause of cancer-related death, due to difficulties in detecting early-stage disease, to its aggressive behaviour, and to poor response to systemic therapy. Therefore, developing strategies for early diagnosis of resectable PC is critical for improving survival. Diabetes mellitus is another major public health problem worldwide. Furthermore, diabetes can represent both a risk factor and a consequence of PC: nowadays, the relationship between these two diseases is considered a high priority for research. New-onset diabetes can be an early manifestation of PC, especially in a thin adult without a family history of diabetes. However, even if targeted screening for patients at higher risk of PC could be a promising approach, this is not recommended in asymptomatic adults with new-onset diabetes, due to the much higher incidence of hyperglycaemia than PC and to the lack of a safe and affordable PC screening test. Prompted by a well-established and productive multidisciplinary cooperation, the Italian Association of Medical Oncology (AIOM), the Italian Medical Diabetologists Association (AMD), the Italian Society of Endocrinology (SIE), and the Italian Society of Pharmacology (SIF) here review available evidence on the mechanisms linking diabetes and PC, addressing the feasibility of screening for early PC in patients with diabetes, and sharing a set of update statements with the aim of providing a state-of-the-art overview and a decision aid tool for daily clinical practice. The incidence of PC is increasing and its prognosis is very poor; therefore, early detection is fundamental. New-onset diabetes may be an early manifestation of PC, often disappearing after its resection. Screening for PC is not currently recommended among people with new-onset diabetes, due to its high incidence. Thin subjects >50 years old at the time of diabetes onset, with sudden weight loss and severe hyperglycaemia are at higher risk. Currently some clinical models are promising for stratifying cancer risk in people with new-onset diabetes.
Collapse
Affiliation(s)
- M Gallo
- Endocrinology and Metabolic Diseases Unit of AO SS Antonio e Biagio e Cesare Arrigo of Alessandria, Alessandria, Italy.
| | - V Adinolfi
- Endocrinology and Diabetology Unit, ASL Verbano Cusio Ossola, Domodossola, Italy
| | - L Morviducci
- Diabetology and Nutrition Unit, Department of Medical Specialities, ASL Roma 1 - S. Spirito Hospital, Rome, Italy
| | - S Acquati
- Endocrinology Unit, Ospedale Pierantoni-Morgagni, Forlì, Italy
| | - E Tuveri
- Diabetology, Endocrinology and Metabolic Diseases Service, ATS Sardegna - ASSL Carbonia-Iglesias, Italy
| | - P Ferrari
- Palliative Care Unit, Istituti Clinici Scientifici Maugeri SPA SB, IRCCS, Pavia, Italy
| | - M C Zatelli
- Section of Endocrinology & Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - A Faggiano
- Endocrinology Unit, Department of Clinical & Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - A Argentiero
- Medical Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - A Natalicchio
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - S D'Oronzo
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - R Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - S Gori
- Oncologia Medica, IRCCS Ospedale Don Calabria-Sacro Cuore di Negrar, Verona, Italy
| | - A Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - M Montagnani
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - G D Beretta
- Medical Oncology Department, Humanitas Gavazzeni, Bergamo, Italy
| | - P Di Bartolo
- Ravenna Diabetes Center, Romagna Diabetes Managed Clinical Network - Romagna Local Health Authority, Ravenna, Italy
| | - N Silvestris
- Medical Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy; Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - F Giorgino
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
39
|
Sugawara T, Ban D, Nishino J, Watanabe S, Maekawa A, Ishikawa Y, Akahoshi K, Ogawa K, Ono H, Kudo A, Tanaka S, Tanabe M. Prediction of early recurrence of pancreatic ductal adenocarcinoma after resection. PLoS One 2021; 16:e0249885. [PMID: 33844700 PMCID: PMC8041173 DOI: 10.1371/journal.pone.0249885] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 03/28/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Even after curative resection, pancreatic ductal adenocarcinoma (PDAC) patients suffer a high rate of recurrence. There is an unmet need to predict which patients will experience early recurrence after resection in order to adjust treatment strategies. METHODS Data of patients with resectable PDAC undergoing surgical resection between January 2005 and September 2018 were reviewed to stratify for early recurrence defined as occurring within 6 months of resection. Preoperative data including demographics, tumor markers, blood immune-inflammatory factors and clinicopathological data were examined. We employed Elastic Net, a sparse modeling method, to construct models predicting early recurrence using these multiple preoperative factors. As a result, seven preoperative factors were selected: age, duke pancreatic monoclonal antigen type 2 value, neutrophil:lymphocyte ratio, systemic immune-inflammation index, tumor size, lymph node metastasis and is peripancreatic invasion. Repeated 10-fold cross-validations were performed, and area under the receiver operating characteristic curve (AUC) and decision curve analysis were used to evaluate the usefulness of the models. RESULTS A total of 136 patients was included in the final analysis, of which 35 (34%) experienced early recurrence. Using Elastic Net, we found that 7 of 14 preoperative factors were useful for the predictive model. The mean AUC of all models constructed in the repeated validation was superior to the standard marker CA 19-9 (0.718 vs 0.657), whereas the AUC of the model constructed from the entire patient cohort was 0.767. Decision curve analysis showed that the models had a higher mean net benefit across the majority of the range of reasonable threshold probabilities. CONCLUSION A model using multiple preoperative factors can improve prediction of early resectable PDAC recurrence.
Collapse
Affiliation(s)
- Toshitaka Sugawara
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daisuke Ban
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Jo Nishino
- Division of Bioinformatics, Research Institute, National Cancer Center Japan, Tokyo, Japan
| | - Shuichi Watanabe
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Aya Maekawa
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshiya Ishikawa
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keiichi Akahoshi
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kosuke Ogawa
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroaki Ono
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsushi Kudo
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Tanaka
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Minoru Tanabe
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
40
|
Krishnaraju VS, Kumar R, Mittal BR, Sharma V, Singh H, Nada R, Bal A, Rohilla M, Singh H, Rana SS. Differentiating benign and malignant pancreatic masses: Ga-68 PSMA PET/CT as a new diagnostic avenue. Eur Radiol 2021; 31:2199-2208. [PMID: 33001304 DOI: 10.1007/s00330-020-07318-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/18/2020] [Accepted: 09/17/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Differentiation of malignant and benign pancreatic lesions on anatomical imaging is difficult in some cases with overlapping features. Prostate-specific membrane antigen (PSMA) is overexpressed during angioneogenesis in many tumors. We aimed to evaluate the PSMA expression in pancreatic lesions to differentiate these lesions and explore the performance of Ga-68 PSMA-PET/CT vis-a-vis F-18 FDG-PET/CT. METHODS Patients with pancreatic lesions on conventional imaging were prospectively recruited. All the patients underwent a whole-body F-18 FDG-PET/CT and a regional abdominal Ga-68 PSMA-PET/CT. Focal tracer uptake (FDG or PSMA) on PET images was considered positive. Histopathology and/or cytopathology were considered the reference standard. RESULTS A total of forty patients (27 males, mean age 55.3 ± 9.8, range 37-71 years) were enrolled. Of these, 19 were diagnosed as malignant on histopathology/cytology. Patients with benign lesions showed no worsening of symptoms for at least 6 months on follow-up. FDG-PET/CT revealed 17 true-positive (TP), 9 false-positive (FP), 12 true-negative (TN), and 2 false-negative (FN) findings, whereas PSMA-PET/CT had 18 TP, 2 FP, 19 TN, and 1 FN finding. The sensitivity, specificity, PPV, NPV, and accuracy for FDG-PET/CT were 89.5%, 57.1%, 65.4%, 85.7%, and 72.5%, respectively, while for PSMA-PET/CT were 94.7%, 90.5%, 90%, 95%, and 92.5%, respectively. ROC curve analysis showed that the SUVmax value of 4.8 on PSMA-PET/CT could predict the malignant potential of a lesion with a specificity of 90.5% and a sensitivity of 84.2%. CONCLUSIONS Ga-68 PSMA-PET/CT imaging helped in establishing a non-invasive pre-operative diagnosis of primary pancreatic malignancy with a higher degree of specificity and accuracy compared with FDG-PET/CT. KEY POINTS • Conventional imaging such as CT and MRI are unable to reliably differentiate localized malignant pancreatic lesion from benign lesions mimicking malignancy such as mass-forming pancreatitis. • FDG PET/CT helps in detecting malignant foci in view of their increased glucose metabolism. However, it may be falsely positive in inflammatory lesions which may occasionally hinder its ability to differentiate between benign and malignant lesions. • Apart from prostatic malignancy, PSMA is overexpressed in neovasculature of many non-prostatic malignancies. The present study highlights that Ga68 PSMA PET/CT performed better in diagnosing malignancy non-invasively than FDG-PET/CT with a higher PPV (90.5% vs. 65.4%) and accuracy (92.5% vs. 72.5%).
Collapse
Affiliation(s)
- Venkata Subramanian Krishnaraju
- Department of Nuclear Medicine and PET/CT, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Rajender Kumar
- Department of Nuclear Medicine and PET/CT, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Bhagwant Rai Mittal
- Department of Nuclear Medicine and PET/CT, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India.
| | - Vishal Sharma
- Department of Gastroenterology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Harjeet Singh
- Department of Surgery, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ritambhra Nada
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Amanjit Bal
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Manish Rohilla
- Department of Cytology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Harmandeep Singh
- Department of Nuclear Medicine and PET/CT, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Surinder S Rana
- Department of Gastroenterology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
41
|
Tseng DSJ, Pranger BK, van Leeuwen MS, Pennings JP, Brosens LA, Mohammad NH, de Meijer VE, van Santvoort HC, Erdmann JI, Molenaar IQ. The Role of CT in Assessment of Extraregional Lymph Node Involvement in Pancreatic and Periampullary Cancer: A Diagnostic Accuracy Study. Radiol Imaging Cancer 2021; 3:e200014. [PMID: 33817647 DOI: 10.1148/rycan.2021200014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 10/15/2020] [Accepted: 11/18/2020] [Indexed: 01/06/2023]
Abstract
Purpose To investigate the diagnostic accuracy of CT in assessing extraregional lymph node metastases in pancreatic head and periampullary cancer. Materials and Methods This prospective observational cohort study was performed at two tertiary hepatopancreatobiliary (HPB) referral centers between March 2013 and December 2014. Patients undergoing pancreatoduodenectomy or bypass surgery with or without palliative radiofrequency ablation were included. Extraregional lymph node involvement was defined as positive lymph nodes in the aortocaval window. Two expert HPB radiologists assessed aortocaval lymph nodes at preoperative CT according to a standardized protocol. All tissue from the aortocaval window was collected intraoperatively. Positive histopathologic finding was the reference standard. Analysis of predictive values and diagnostic accuracy was performed. Results A total of 198 consecutive patients (mean age, 66 years; range, 39-86 years; 105 men) with pancreatic head or periampullary carcinoma were included. In 70% of patients, a pancreatoduodenectomy was performed, 4% underwent total pancreatectomy, 4% underwent radiofrequency ablation, and 22% underwent bypass surgery. Forty-four patients (22%) had histologically positive aortocaval lymph nodes. Negative predictive value of CT in assessing aortocaval lymph nodes was 80% for both observers, and positive predictive value was 31%-33%. Overall diagnostic accuracy was 69%-70%. Conclusion CT has a low diagnostic accuracy in assessing extraregional lymph node metastases in patients suspected of having pancreatic or periampullary cancer.Keywords: CT, Abdomen/GI, Pancreas, Oncology© RSNA, 2021.
Collapse
Affiliation(s)
- Dorine S J Tseng
- Department of Surgery, University Medical Center Utrecht and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Heidelberglaan 100, HG G04.228, PO Box 85500, 3508 GA, Utrecht, the Netherlands (D.S.J.T., H.C.v.S., I.Q.M.); University of Groningen and University Medical Center Groningen, Division of Hepatopancreatobiliary Surgery and Liver Transplantation, Groningen, the Netherlands (B.K.P., V.E.d.M., J.I.E.); Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands (M.S.v.L.); University of Groningen and University Medical Center Groningen, Department of Radiology, Groningen, the Netherlands (J.P.P.); Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands (L.A.B.); Department of Medical Oncology, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands (N.H.M.)
| | - Bobby K Pranger
- Department of Surgery, University Medical Center Utrecht and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Heidelberglaan 100, HG G04.228, PO Box 85500, 3508 GA, Utrecht, the Netherlands (D.S.J.T., H.C.v.S., I.Q.M.); University of Groningen and University Medical Center Groningen, Division of Hepatopancreatobiliary Surgery and Liver Transplantation, Groningen, the Netherlands (B.K.P., V.E.d.M., J.I.E.); Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands (M.S.v.L.); University of Groningen and University Medical Center Groningen, Department of Radiology, Groningen, the Netherlands (J.P.P.); Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands (L.A.B.); Department of Medical Oncology, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands (N.H.M.)
| | - Maarten S van Leeuwen
- Department of Surgery, University Medical Center Utrecht and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Heidelberglaan 100, HG G04.228, PO Box 85500, 3508 GA, Utrecht, the Netherlands (D.S.J.T., H.C.v.S., I.Q.M.); University of Groningen and University Medical Center Groningen, Division of Hepatopancreatobiliary Surgery and Liver Transplantation, Groningen, the Netherlands (B.K.P., V.E.d.M., J.I.E.); Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands (M.S.v.L.); University of Groningen and University Medical Center Groningen, Department of Radiology, Groningen, the Netherlands (J.P.P.); Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands (L.A.B.); Department of Medical Oncology, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands (N.H.M.)
| | - Jan Pieter Pennings
- Department of Surgery, University Medical Center Utrecht and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Heidelberglaan 100, HG G04.228, PO Box 85500, 3508 GA, Utrecht, the Netherlands (D.S.J.T., H.C.v.S., I.Q.M.); University of Groningen and University Medical Center Groningen, Division of Hepatopancreatobiliary Surgery and Liver Transplantation, Groningen, the Netherlands (B.K.P., V.E.d.M., J.I.E.); Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands (M.S.v.L.); University of Groningen and University Medical Center Groningen, Department of Radiology, Groningen, the Netherlands (J.P.P.); Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands (L.A.B.); Department of Medical Oncology, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands (N.H.M.)
| | - Lodewijk A Brosens
- Department of Surgery, University Medical Center Utrecht and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Heidelberglaan 100, HG G04.228, PO Box 85500, 3508 GA, Utrecht, the Netherlands (D.S.J.T., H.C.v.S., I.Q.M.); University of Groningen and University Medical Center Groningen, Division of Hepatopancreatobiliary Surgery and Liver Transplantation, Groningen, the Netherlands (B.K.P., V.E.d.M., J.I.E.); Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands (M.S.v.L.); University of Groningen and University Medical Center Groningen, Department of Radiology, Groningen, the Netherlands (J.P.P.); Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands (L.A.B.); Department of Medical Oncology, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands (N.H.M.)
| | - Nadja Haj Mohammad
- Department of Surgery, University Medical Center Utrecht and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Heidelberglaan 100, HG G04.228, PO Box 85500, 3508 GA, Utrecht, the Netherlands (D.S.J.T., H.C.v.S., I.Q.M.); University of Groningen and University Medical Center Groningen, Division of Hepatopancreatobiliary Surgery and Liver Transplantation, Groningen, the Netherlands (B.K.P., V.E.d.M., J.I.E.); Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands (M.S.v.L.); University of Groningen and University Medical Center Groningen, Department of Radiology, Groningen, the Netherlands (J.P.P.); Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands (L.A.B.); Department of Medical Oncology, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands (N.H.M.)
| | - Vincent E de Meijer
- Department of Surgery, University Medical Center Utrecht and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Heidelberglaan 100, HG G04.228, PO Box 85500, 3508 GA, Utrecht, the Netherlands (D.S.J.T., H.C.v.S., I.Q.M.); University of Groningen and University Medical Center Groningen, Division of Hepatopancreatobiliary Surgery and Liver Transplantation, Groningen, the Netherlands (B.K.P., V.E.d.M., J.I.E.); Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands (M.S.v.L.); University of Groningen and University Medical Center Groningen, Department of Radiology, Groningen, the Netherlands (J.P.P.); Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands (L.A.B.); Department of Medical Oncology, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands (N.H.M.)
| | - Hjalmar C van Santvoort
- Department of Surgery, University Medical Center Utrecht and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Heidelberglaan 100, HG G04.228, PO Box 85500, 3508 GA, Utrecht, the Netherlands (D.S.J.T., H.C.v.S., I.Q.M.); University of Groningen and University Medical Center Groningen, Division of Hepatopancreatobiliary Surgery and Liver Transplantation, Groningen, the Netherlands (B.K.P., V.E.d.M., J.I.E.); Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands (M.S.v.L.); University of Groningen and University Medical Center Groningen, Department of Radiology, Groningen, the Netherlands (J.P.P.); Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands (L.A.B.); Department of Medical Oncology, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands (N.H.M.)
| | - Joris I Erdmann
- Department of Surgery, University Medical Center Utrecht and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Heidelberglaan 100, HG G04.228, PO Box 85500, 3508 GA, Utrecht, the Netherlands (D.S.J.T., H.C.v.S., I.Q.M.); University of Groningen and University Medical Center Groningen, Division of Hepatopancreatobiliary Surgery and Liver Transplantation, Groningen, the Netherlands (B.K.P., V.E.d.M., J.I.E.); Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands (M.S.v.L.); University of Groningen and University Medical Center Groningen, Department of Radiology, Groningen, the Netherlands (J.P.P.); Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands (L.A.B.); Department of Medical Oncology, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands (N.H.M.)
| | - I Quintus Molenaar
- Department of Surgery, University Medical Center Utrecht and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Heidelberglaan 100, HG G04.228, PO Box 85500, 3508 GA, Utrecht, the Netherlands (D.S.J.T., H.C.v.S., I.Q.M.); University of Groningen and University Medical Center Groningen, Division of Hepatopancreatobiliary Surgery and Liver Transplantation, Groningen, the Netherlands (B.K.P., V.E.d.M., J.I.E.); Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands (M.S.v.L.); University of Groningen and University Medical Center Groningen, Department of Radiology, Groningen, the Netherlands (J.P.P.); Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands (L.A.B.); Department of Medical Oncology, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands (N.H.M.)
| |
Collapse
|
42
|
Yamada M, Sugiura T, Okamura Y, Ito T, Yamamoto Y, Ashida R, Ohgi K, Aramaki T, Endo M, Uesaka K. Clinical Implication of Node-negative Resectable Pancreatic Cancer. Ann Surg Oncol 2021; 28:2257-2264. [PMID: 33452602 DOI: 10.1245/s10434-020-09543-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 11/05/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Lymph node metastasis is one of the strongest prognostic factors of pancreatic cancer. However, the clinical implication of pathologically node-negative pancreatic cancer (pN0-PC) has not been fully investigated. METHODS Patients who underwent surgical resection for radiologically resectable pancreatic cancer between 2002 and 2018 were included in this study. A clinicopathological examination focusing on pN0-PC was performed. RESULTS Of all 533 patients, 155 (29.1%) were diagnosed with pN0-PC and 378 (70.9%) were diagnosed with node-positive pancreatic cancer (pN1/2-PC). The 5-year survival rates of patients with pN0-PC and pN1/2-PC were 57.1% and 25.0%, respectively (p < 0.001). A multivariate analysis revealed six prognostic factors in pN0-PC: age ≥ 70 years, nonadministration of adjuvant chemotherapy, anterior serosal invasion, nerve plexus invasion, and microscopic lymphatic and venous invasions. The 5-year survival rates of patients who had pN0-PC with 0-1 risk factor, with 2-3 risk factors, and with 4-6 risk factors were 87.6%, 47.9%, and 16.4%, respectively. Survival of patients who had pN0-PC with 4-6 risk factors was comparable to that of pN1/2 patients. The diagnostic capability of metastasis-negative lymph node was unsatisfactory, with a predictive value of < 43%. CONCLUSIONS Although the prognosis of patients with pN0-PC was better than that of patients with pN1/2-PC, it is not satisfactory. Survival of patients who had pN0-PC with 0-1 risk factors was extremely favorable; however, survival of patients who had pN0-PC with 4-6 risk factors was similar to those with pN1/2-PC.
Collapse
Affiliation(s)
- Mihoko Yamada
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Teiichi Sugiura
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan.
| | - Yukiyasu Okamura
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Takaaki Ito
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Yusuke Yamamoto
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Ryo Ashida
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Katsuhisa Ohgi
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Takeshi Aramaki
- Division of Diagnostic Radiology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Masahiro Endo
- Division of Diagnostic Radiology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Katsuhiko Uesaka
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
43
|
|
44
|
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
|
45
|
Aronen A, Aittoniemi J, Huttunen R, Nikkola A, Rinta-Kiikka I, Nikkola J, Limnell O, Nordback I, Sand J, Laukkarinen J. Plasma suPAR may help to distinguish between chronic pancreatitis and pancreatic cancer. Scand J Gastroenterol 2021; 56:81-85. [PMID: 33245246 DOI: 10.1080/00365521.2020.1849383] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES SuPAR (soluble urokinase-type plasminogen activator receptor) is a biomarker reflecting the inflammatory state of the human body. Earlier studies suggest that urinary suPAR/creatinine ratio levels are elevated in chronic pancreatitis (CP), and that plasma suPAR (P-suPAR) level is elevated in pancreatic cancer (PC). Our aim was to study the levels of P-suPAR in CP in a long-term prospective follow-up setting to explore the possibility of distinguishing between PC and CP. MATERIALS AND METHODS Two patient groups were compared. The first group included 83 patients who were prospectively followed up after their first acute alcohol-induced pancreatitis (AAP) for median 7.0 (range 0.3-9.8) years. Twelve patients in this group developed CP during follow-up, and two patients were further excluded from the CP cohort. The second group consisted of 25 patients operated on for suspicion of pancreatic malignancy and final pathological diagnosis of PC. P-suPAR levels were measured and compared within and between these groups. RESULTS P-suPAR levels remained low during follow-up despite the development of CP. P-suPAR was significantly higher in PC patients [median 3.7 (IQR 3.1-4.4) ng/mL] than in CP patients [2.6 (1.8-3.6) ng/mL]; p = .014. A cutoff value of 2.8 ng/mL resulted from a ROC curve with area under curve (AUC) of 0.79 (95% CI 0.61-0.97), p = .009 in differentiation between PC and CP with a sensitivity and a specificity of 88% and 70% respectively. CONCLUSION P-suPAR is higher in patients with PC than in patients with CP, and it could thus be used in differentiating between PC and CP.
Collapse
Affiliation(s)
- Anu Aronen
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
| | | | - Reetta Huttunen
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Anssi Nikkola
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland.,Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Irina Rinta-Kiikka
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Department of Radiology, Tampere University Hospital, Tampere, Finland
| | - Jussi Nikkola
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
| | - Olli Limnell
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Isto Nordback
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
| | - Juhani Sand
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
| | - Johanna Laukkarinen
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland.,Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
46
|
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]
|
47
|
Impact of F-18 Fluorodeoxyglucose PET/CT and PET/MRI on Initial Staging and Changes in Management of Pancreatic Ductal Adenocarcinoma: A Systemic Review and Meta-Analysis. Diagnostics (Basel) 2020; 10:diagnostics10110952. [PMID: 33202682 PMCID: PMC7696716 DOI: 10.3390/diagnostics10110952] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023] Open
Abstract
A systemic review and meta-analysis were conducted to investigate the diagnostic ability for staging and impact on management of F-18 fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) and PET/magnetic resonance imaging (MRI) in patients with pancreatic ductal adenocarcinoma. A comprehensive search was performed in four databases to retrieve studies of pancreatic ductal adenocarcinoma patients that have reported the diagnostic ability of FDG PET/CT and PET/MRI for detecting metastasis and the proportion of patients whose management was changed by its results. The sensitivity and specificity for detecting metastasis and the proportion of patients with management changes were pooled using a random-effects model. A total of 10 studies were included. The pooled sensitivity and specificity for detecting lymph node metastasis were 0.55 and 0.94, respectively, while the pooled sensitivity and specificity for detecting distant metastasis were 0.80 and 1.00, respectively. The areas under the summarized receiver operating characteristic curves for detecting lymph node and distant metastasis were 0.88 and 0.92, respectively. The pooled proportion of patients with management changes was 19%. FDG PET/CT and PET/MRI showed high diagnostic accuracy for detecting lymph node and distant metastasis in pancreatic ductal adenocarcinoma patients, and the use of these imaging tools led to management changes in a significant portion of these patients.
Collapse
|
48
|
Que R, Chen Y, Tao Z, Ge B, Li M, Fu Z, Li Y. Diffusion-weighted MRI versus FDG-PET/CT for diagnosing pancreatic cancer: an indirect comparison meta-analysis. Acta Radiol 2020; 61:1473-1483. [PMID: 32148066 DOI: 10.1177/0284185120907246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Fluorodeoxyglucose positron emission tomography (FDG-PET)/computed tomography (CT) and diffusion-weighted magnetic resonance imaging (DWI or DW-MRI) are tools for the diagnosis of pancreatic cancer. However, comparison of their diagnostic performance remains unknown. PURPOSE To indirectly compare the diagnostic value of DWI and FDG-PET/CT in the detection of pancreatic cancer. MATERIAL AND METHODS A literature search of PubMed, Embase, and Cochrane Library electronic databases for articles published through May 2018 yielded 875 articles. For the meta-analysis, we included 26 studies evaluating the efficacy of DWI and FDG-PET/CT for determining pancreatic cancer with a total of 1377 patients. QUADAS (Quality Assessment of Diagnostic Accuracy Studies) was used to assess the study quality. Sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and the area under the receiver operating characteristic curves (AUC) with their 95% confidence intervals were calculated for each individual study. RESULTS There were no significant differences between DWI and FDG-PET/CT for sensitivity, specificity, PLR, NLR, or DOR, while DWI AUC was higher than that of FDG-PET/CT for the detection of pancreatic cancer. CONCLUSION The diagnostic value of both DWI and FDG-PET/CT were comparable and, hence, both techniques seem to be equally useful tools for the diagnosis of pancreatic cancer.
Collapse
Affiliation(s)
- Renye Que
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
- Department of Gastroenterology, Shanghai TCM Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Yirong Chen
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Zhihui Tao
- Department of Oncology, Jiading Hospital of Traditional Chinese Medicine, Shanghai, PR China
| | - Bingjing Ge
- Department of Gastroenterology, Shanghai TCM Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Miaohua Li
- Department of Gastroenterology, Shanghai TCM Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Zhiquan Fu
- Department of Gastroenterology, Shanghai TCM Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Yong Li
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| |
Collapse
|
49
|
Röhrich M, Naumann P, Giesel FL, Choyke PL, Staudinger F, Wefers A, Liew DP, Kratochwil C, Rathke H, Liermann J, Herfarth K, Jäger D, Debus J, Haberkorn U, Lang M, Koerber SA. Impact of 68Ga-FAPI PET/CT Imaging on the Therapeutic Management of Primary and Recurrent Pancreatic Ductal Adenocarcinomas. J Nucl Med 2020; 62:779-786. [PMID: 33097632 DOI: 10.2967/jnumed.120.253062] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/29/2020] [Indexed: 01/13/2023] Open
Abstract
Pancreatic ductal carcinoma (PDAC) is a highly lethal cancer, and early detection and accurate staging are critical to prolonging survival. PDAC typically has a prominent stroma including cancer-associated fibroblasts that express fibroblast activation protein (FAP). FAP is a new target molecule for PET imaging of various tumors. In this retrospective study, we describe the clinical impact of PET/CT imaging using 68Ga-labeled FAP-inhibitors (68Ga-FAPI PET/CT) in 19 patients with PDAC (7 primary, 12 progressive/recurrent). Methods: All patients underwent contrast-enhanced CT (ceCT) for TNM staging before 68Ga-FAPI PET/CT imaging. PET scans were acquired 60 min after administration of 150-250 MBq of 68Ga-labeled FAP-specific tracers. To characterize 68Ga-FAPI uptake over time, additional scans after 10 or 180 min were acquired in 6 patients. SUVmax and SUVmean values of PDAC manifestations and healthy organs were analyzed. The tumor burden according to 68Ga-FAPI PET/CT was compared with TNM staging based on ceCT and changes in oncologic management were recorded. Results: Compared with ceCT, 68Ga-FAPI PET/CT results led to changes in TNM staging in 10 of 19 patients. Eight of 12 patients with recurrent/progressive disease were upstaged, 1 was downstaged, and 3 had no change. In newly diagnosed PDAC, 1 of 7 patients was upstaged, and the staging of 6 patients did not change. Changes in oncologic management occurred in 7 patients. Markedly elevated uptake of 68Ga-FAPI in PDAC manifestations after 1 h was seen in most cases. Differentiation from pancreatitis based on static imaging 1 h after injection was challenging. With respect to imaging after multiple time points, PDAC and pancreatitis showed a trend for differential uptake kinetics. Conclusion: 68Ga-FAPI PET/CT led to restaging in half of the patients with PDAC and most patients with recurrent disease compared with standard of care imaging. The clinical value of 68Ga-FAPI PET/CT should be further investigated.
Collapse
Affiliation(s)
- Manuel Röhrich
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Patrick Naumann
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor diseases (NCT), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Frederik L Giesel
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), partner site Heidelberg, Germany
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Fabian Staudinger
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Annika Wefers
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dawn P Liew
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Hendrik Rathke
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Jakob Liermann
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor diseases (NCT), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Klaus Herfarth
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor diseases (NCT), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology and Internal Medicine Virgin Islands, National Center for Tumor Diseases, University Hospital Heidelberg, Germany.,Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor diseases (NCT), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,German Cancer Consortium (DKTK), partner site Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), partner site Heidelberg, Germany.,Clinical Cooperation Unit, Department of Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research DZL, Heidelberg, Germany; and
| | - Matthias Lang
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Department of Surgery, Heidelberg University Hospital, Heidelberg, GermanyMember of the German Center for Lung Research DZL, Heidelberg, Germany
| | - Stefan A Koerber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor diseases (NCT), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| |
Collapse
|
50
|
Non-coding RNA biomarkers in pancreatic ductal adenocarcinoma. Semin Cancer Biol 2020; 75:153-168. [PMID: 33049362 DOI: 10.1016/j.semcancer.2020.10.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/20/2020] [Accepted: 10/02/2020] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies, which is usually diagnosed at an advanced stage. The late disease diagnosis, the limited availability of effective therapeutic interventions and lack of robust diagnostic biomarkers, are some of the primary reasons for the dismal 5-year survival rates (∼8%) in patients with PDAC. The pancreatic cancer develops through accumulation of a series of genomic and epigenomic alterations which lead to the transformation of normal pancreatic epithelium into an invasive carcinoma - a process that can take up to 15-20 years to develop, from the occurrence of first initiating mutational event. These facts highlight a unique window of opportunity for the earlier detection of PDAC, which could allow timely disease interception and improvement in the overall survival outcomes in patients suffering from this fatal malignancy. Non-coding RNAs (ncRNAs) have been recognized to play a central role in PDAC pathogenesis and are emerging as attractive candidates for biomarker development in various cancers, including PDAC. More specifically, the ncRNAs play a pivotal role in PDAC biology as they affect tumor growth, migration, and invasion by regulating cellular processes including cell cycle, apoptosis, and epithelial-mesenchymal transition. In this review, we focus on three types of well-established ncRNAs - microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) - and discuss their potential as diagnostic, prognostic and predictive biomarkers in PDAC.
Collapse
|