1
|
Ahn J, Song YS, Kim B, Yang S, Jung K, Lee JC, Kim J, Hwang JH. Prognostic Value of the Metabolic Response on Serial 18F-FDG PET/CT in Pancreatic Cancer. Gut Liver 2025; 19:462-472. [PMID: 40051310 PMCID: PMC12070205 DOI: 10.5009/gnl240458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/24/2024] [Accepted: 12/29/2024] [Indexed: 05/14/2025] Open
Abstract
Background/Aims The prognostic value of serial 18F-fluorodeoxyglucose positron emission tomography/ computed tomography (18F-FDG PET/CT) for patients with borderline resectable or locally advanced pancreatic cancer who undergo conversion surgery or continue chemotherapy without surgery has not been well-established. Methods A retrospective analysis of patients with pancreatic ductal adenocarcinoma was conducted at Seoul National University Bundang Hospital between March 2013 and February 2022. Patients underwent PET/CT at baseline and subsequent radiologic evaluations following chemotherapy. Changes in the maximum standardized uptake value (SUVmax), mean standardized uptake value (SUVmean), metabolic tumor volume, and total lesion glycolysis were analyzed. Based on their treatment regimens, patients were stratified into the conversion surgery group or nonconversion surgery group. Survival outcomes and various clinical factors were assessed. Results Among 121 patients, 52 underwent conversion surgery, and 69 continued to receive chemotherapy without surgery. A significant reduction in the SUVmax was correlated with prolonged recurrence-free survival and overall survival in the conversion surgery group. Confirmation of a pathologic response indicated a significant association between reductions in the SUVmax and positive outcomes. Reductions in the metabolic tumor volume and total lesion glycolysis were associated with improved progression-free survival and overall survival in the nonconversion surgery group. Conclusions Serial PET/CT scans demonstrated prognostic value in pancreatic ductal adenocarcinoma patients, revealing distinct correlations in the conversion surgery group and nonconversion surgery group.
Collapse
Affiliation(s)
- Jinwoo Ahn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Yoo Sung Song
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Bomi Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Soomin Yang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Kwangrok Jung
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jong-Chan Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
2
|
Javed AA, Habib A, Mahmud O, Fatimi AS, Grewal M, Mughal N, He J, Wolfgang CL, Daamen L, Besselink MG. Prognostic factors in localized pancreatic ductal adenocarcinoma after neoadjuvant therapy and resection: a systematic review and meta-analysis. J Natl Cancer Inst 2025; 117:840-867. [PMID: 39563429 DOI: 10.1093/jnci/djae294] [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: 09/26/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Prognostic markers for overall survival in resected pancreatic ductal adenocarcinoma are well established but remain unclear following neoadjuvant therapy. This systematic review and meta-analysis aimed to determine factors associated with overall survival following neoadjuvant therapy in resected pancreatic ductal adenocarcinoma. METHODS The PubMed, Embase, Scopus, Web of Science, and Cochrane CENTRAL databases were systematically searched from January 2010 until May 2024. Studies that reported univariable and multivariable hazard ratios were included if patients underwent neoadjuvant therapy and resection for localized pancreatic ductal adenocarcinoma. Study quality assessment was performed using the Newcastle-Ottawa scale. Meta-analysis was performed using generic inverse-variance random-effects models. RESULTS Among 2208 unique articles identified by the search, 92 were included in the meta-analysis. Of these, 85 were of "good" and 7 of "poor" quality. The neoadjuvant therapy regimen was described in 84 studies of which 62 included patients treated with FOLFIRINOX. Margin status, nodal disease, American Joint Committee on Cancer (AJCC) T-stage, and normalization of cancer antigen 19-9 (CA19-9) after neoadjuvant therapy were prognostic for overall survival, whereas age, sex, perineural invasion, baseline tumor size, and baseline CA19-9 were not. The test for subgroup differences between ypN substages was not statistically significant in the multivariable model. Neoadjuvant FOLFIRINOX was associated with better survival than other regimens. CONCLUSIONS This meta-analysis identified margin status, nodal disease, AJCC T-stage, and normalization of CA19-9 after neoadjuvant therapy as prognostic factors for overall survival in patients with resected localized pancreatic ductal adenocarcinoma following neoadjuvant therapy.
Collapse
Affiliation(s)
- Ammar A Javed
- New York University Langone Health, New York University Grossman School of Medicine, New York City, NY 10016, United States
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam 1007 MB, the Netherlands
- Cancer Center Amsterdam, Amsterdam 1007 MB, the Netherlands
| | - Alyssar Habib
- New York University Langone Health, New York University Grossman School of Medicine, New York City, NY 10016, United States
| | - Omar Mahmud
- New York University Langone Health, New York University Grossman School of Medicine, New York City, NY 10016, United States
- Medical College, Aga Khan University, Karachi 74800, Pakistan
| | - Asad Saulat Fatimi
- New York University Langone Health, New York University Grossman School of Medicine, New York City, NY 10016, United States
- Medical College, Aga Khan University, Karachi 74800, Pakistan
| | - Mahip Grewal
- New York University Langone Health, New York University Grossman School of Medicine, New York City, NY 10016, United States
| | - Nabiha Mughal
- New York University Langone Health, New York University Grossman School of Medicine, New York City, NY 10016, United States
| | - Jin He
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21205, United States
| | - Christopher L Wolfgang
- New York University Langone Health, New York University Grossman School of Medicine, New York City, NY 10016, United States
| | - Lois Daamen
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht 3508 GA, the Netherlands
| | - Marc G Besselink
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam 1007 MB, the Netherlands
- Cancer Center Amsterdam, Amsterdam 1007 MB, the Netherlands
| |
Collapse
|
3
|
Chuong MD, Ashman J, Jethwa K, Kharofa J, Kim H, Koay E, Ludmir E, Miller E, Nelson B, Reyngold M, Sanford N, Chang D. Moving From the Background Toward the Spotlight: A Critical Review of Radiation Therapy for Locally Advanced Pancreas Cancer. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00162-2. [PMID: 40032056 DOI: 10.1016/j.ijrobp.2025.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/31/2025] [Accepted: 02/16/2025] [Indexed: 03/05/2025]
Abstract
Radiation therapy (RT) for locally advanced pancreatic cancer (LAPC) continues to be controversial. Advances in both systemic therapy and RT techniques have changed the landscape of LAPC management in recent years. Clinical outcomes of ablative RT have been encouraging, and randomized clinical trials may clarify the role of RT for LAPC. We present a contemporary critical review of key aspects regarding optimal patient selection, radiation dose escalation techniques, novel radiosensitizers and radioprotectors, and treatment response assessment for LAPC.
Collapse
Affiliation(s)
- Michael D Chuong
- Department of Radiation Oncology, Miami Cancer Institute, Miami, Florida.
| | - Jonathan Ashman
- Department of Radiation Oncology, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Krishan Jethwa
- Department of Radiation Oncology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Jordan Kharofa
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Hyun Kim
- Department of Radiation Oncology, Washington University in St. Louis, Missouri.
| | - Eugene Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ethan Ludmir
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eric Miller
- Department of Radiation Oncology, Ohio State University, Columbus, Ohio
| | - Bailey Nelson
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Marsha Reyngold
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nina Sanford
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Daniel Chang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
4
|
Antony A, Mukherjee S, Bi Y, Collisson EA, Nagaraj M, Murlidhar M, Wallace MB, Goenka AH. AI-Driven insights in pancreatic cancer imaging: from pre-diagnostic detection to prognostication. Abdom Radiol (NY) 2024:10.1007/s00261-024-04775-x. [PMID: 39738571 DOI: 10.1007/s00261-024-04775-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related deaths in the United States, largely due to its poor five-year survival rate and frequent late-stage diagnosis. A significant barrier to early detection even in high-risk cohorts is that the pancreas often appears morphologically normal during the pre-diagnostic phase. Yet, the disease can progress rapidly from subclinical stages to widespread metastasis, undermining the effectiveness of screening. Recently, artificial intelligence (AI) applied to cross-sectional imaging has shown significant potential in identifying subtle, early-stage changes in pancreatic tissue that are often imperceptible to the human eye. Moreover, AI-driven imaging also aids in the discovery of prognostic and predictive biomarkers, essential for personalized treatment planning. This article uniquely integrates a critical discussion on AI's role in detecting visually occult PDAC on pre-diagnostic imaging, addresses challenges of model generalizability, and emphasizes solutions like standardized datasets and clinical workflows. By focusing on both technical advancements and practical implementation, this article provides a forward-thinking conceptual framework that bridges current gaps in AI-driven PDAC research.
Collapse
Affiliation(s)
- Ajith Antony
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Yan Bi
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Eric A Collisson
- Department of Medical Oncology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Madhu Nagaraj
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Michael B Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Ajit H Goenka
- Department of Radiology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
5
|
Steadman JA, Sultan A, Day CN, Parish MA, Warner SG, Kendrick ML, Truty MJ, Jin Z, Thiels CA. Impact of proton pump inhibitors on pathologic response rates following fluoropyrimidine-based neoadjuvant chemotherapy in pancreatic cancer patients. J Surg Oncol 2024; 130:1634-1642. [PMID: 39257300 DOI: 10.1002/jso.27837] [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: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND Proton pump inhibitors (PPIs) negatively impact fluoropyrimidine-based chemotherapy efficacy in colorectal cancer. This study assessed PPI impact on major pathologic response (mPR) rates of pancreatic adenocarcinoma (PDAC) patients receiving fluoropyrimidine-based chemotherapy. METHODS An institutional retrospective review of resected PDAC patients receiving neoadjuvant fluoropyrimidine-based chemotherapy (98% FOLFIRINOX) from 2011 to 2021 was conducted. Outcomes were stratified by use or nonuse of PPIs within 6 months of neoadjuvant chemotherapy initiation. Primary outcome was mPR defined as complete or near complete response. RESULTS Among 540 patients included, the median age was 64 (IQR: 60-70) years, 297 (55%) were male, and 202 (37%) were PPI users. 170 (31%) patients had mPR with similar rates among PPI users and nonusers (29% vs. 33%, p = 0.38). No difference in mPR was seen between PPI users and nonusers receiving chemoradiation (35% vs. 36%, p = 0.89) or ≥8 cycles of NAC (33% vs. 36%, p = 0.55). Median OS for PPI users was 30.9 versus 31.7 months for nonusers (p = 0.62). On multivariable analysis, PPI therapy was not associated with decreased survival. CONCLUSION PPI usage did not significantly influence mPR or OS following neoadjuvant fluoropyrimidine-based chemotherapy in resected PDAC patients. Further analysis of all patients, not just those who underwent resection, is required.
Collapse
Affiliation(s)
- Jessica A Steadman
- Division of Hepatobiliary & Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ahmer Sultan
- Division of Hepatobiliary & Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Courtney N Day
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Marie A Parish
- Division of Pharmacy Cancer Care, Mayo Clinic, Rochester, Minnesota, USA
| | - Susanne G Warner
- Division of Hepatobiliary & Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael L Kendrick
- Division of Hepatobiliary & Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark J Truty
- Division of Hepatobiliary & Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Zhaohui Jin
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Cornelius A Thiels
- Division of Hepatobiliary & Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
6
|
Liu W, Zhang B, Liu T, Jiang J, Liu Y. Artificial Intelligence in Pancreatic Image Analysis: A Review. SENSORS (BASEL, SWITZERLAND) 2024; 24:4749. [PMID: 39066145 PMCID: PMC11280964 DOI: 10.3390/s24144749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Pancreatic cancer is a highly lethal disease with a poor prognosis. Its early diagnosis and accurate treatment mainly rely on medical imaging, so accurate medical image analysis is especially vital for pancreatic cancer patients. However, medical image analysis of pancreatic cancer is facing challenges due to ambiguous symptoms, high misdiagnosis rates, and significant financial costs. Artificial intelligence (AI) offers a promising solution by relieving medical personnel's workload, improving clinical decision-making, and reducing patient costs. This study focuses on AI applications such as segmentation, classification, object detection, and prognosis prediction across five types of medical imaging: CT, MRI, EUS, PET, and pathological images, as well as integrating these imaging modalities to boost diagnostic accuracy and treatment efficiency. In addition, this study discusses current hot topics and future directions aimed at overcoming the challenges in AI-enabled automated pancreatic cancer diagnosis algorithms.
Collapse
Affiliation(s)
- Weixuan Liu
- Sydney Smart Technology College, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China; (W.L.); (B.Z.)
| | - Bairui Zhang
- Sydney Smart Technology College, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China; (W.L.); (B.Z.)
| | - Tao Liu
- School of Mathematics and Statistics, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China;
| | - Juntao Jiang
- College of Control Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yong Liu
- College of Control Science and Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
7
|
Jeong B, Oh M, Lee SS, Kim N, Kim JS, Lee W, Kim SC, Kim HJ, Kim JH, Byun JH. Predicting Recurrence-Free Survival After Upfront Surgery in Resectable Pancreatic Ductal Adenocarcinoma: A Preoperative Risk Score Based on CA 19-9, CT, and 18F-FDG PET/CT. Korean J Radiol 2024; 25:644-655. [PMID: 38942458 PMCID: PMC11214925 DOI: 10.3348/kjr.2023.1235] [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: 12/12/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 06/30/2024] Open
Abstract
OBJECTIVE To develop and validate a preoperative risk score incorporating carbohydrate antigen (CA) 19-9, CT, and fluorine-18-fluorodeoxyglucose (18F-FDG) PET/CT variables to predict recurrence-free survival (RFS) after upfront surgery in patients with resectable pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS Patients with resectable PDAC who underwent upfront surgery between 2014 and 2017 (development set) or between 2018 and 2019 (test set) were retrospectively evaluated. In the development set, a risk-scoring system was developed using the multivariable Cox proportional hazards model, including variables associated with RFS. In the test set, the performance of the risk score was evaluated using the Harrell C-index and compared with that of the postoperative pathological tumor stage. RESULTS A total of 529 patients, including 335 (198 male; mean age ± standard deviation, 64 ± 9 years) and 194 (103 male; mean age, 66 ± 9 years) patients in the development and test sets, respectively, were evaluated. The risk score included five variables predicting RFS: tumor size (hazard ratio [HR], 1.29 per 1 cm increment; P < 0.001), maximal standardized uptake values of tumor ≥ 5.2 (HR, 1.29; P = 0.06), suspicious regional lymph nodes (HR, 1.43; P = 0.02), possible distant metastasis on 18F-FDG PET/CT (HR, 2.32; P = 0.03), and CA 19-9 (HR, 1.02 per 100 U/mL increment; P = 0.002). In the test set, the risk score showed good performance in predicting RFS (C-index, 0.61), similar to that of the pathologic tumor stage (C-index, 0.64; P = 0.17). CONCLUSION The proposed risk score based on preoperative CA 19-9, CT, and 18F-FDG PET/CT variables may have clinical utility in selecting high-risk patients with resectable PDAC.
Collapse
Affiliation(s)
- Boryeong Jeong
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Minyoung Oh
- Department of Nuclear Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Seung Soo Lee
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea.
| | - Nayoung Kim
- Department of Clinical Epidemiology and Biostatistics, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jae Seung Kim
- Department of Nuclear Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Woohyung Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Song Cheol Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Hyoung Jung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jin Hee Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jae Ho Byun
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
8
|
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
|
9
|
Koti S, Demyan L, Deutsch G, Weiss M. Surgery for Oligometastatic Pancreatic Cancer: Defining Biologic Resectability. Ann Surg Oncol 2024; 31:4031-4041. [PMID: 38502293 PMCID: PMC11076395 DOI: 10.1245/s10434-024-15129-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/17/2024] [Indexed: 03/21/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is most often metastatic at diagnosis. As systemic therapy continues to improve alongside advanced surgical techniques, the focus has shifted toward defining biologic, rather than technical, resectability. Several centers have reported metastasectomy for oligometastatic PDAC, yet the indications and potential benefits remain unclear. In this review, we attempt to define oligometastatic disease in PDAC and to explore the rationale for metastasectomy. We evaluate the existing evidence for metastasectomy in liver, peritoneum, and lung individually, assessing the safety and oncologic outcomes for each. Furthermore, we explore contemporary biomarkers of biological resectability in oligometastatic PDAC, including radiographic findings, biochemical markers (such as CA 19-9 and CEA), inflammatory markers (including neutrophil-to-lymphocyte ratio, C-reactive protein, and scoring indices), and liquid biopsy techniques. With careful consideration of existing data, we explore the concept of biologic resectability in guiding patient selection for metastasectomy in PDAC.
Collapse
Affiliation(s)
- Shruti Koti
- Department of General Surgery, Northwell Health, Queens, NY, USA.
- Northwell Health Cancer Institute, Northwell Health, New Hyde Park, NY, USA.
| | - Lyudmyla Demyan
- Department of General Surgery, Northwell Health, Queens, NY, USA
| | - Gary Deutsch
- Northwell Health Cancer Institute, Northwell Health, New Hyde Park, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Matthew Weiss
- Northwell Health Cancer Institute, Northwell Health, New Hyde Park, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| |
Collapse
|
10
|
Bilreiro C, Andrade L, Santiago I, Marques RM, Matos C. Imaging of pancreatic ductal adenocarcinoma - An update for all stages of patient management. Eur J Radiol Open 2024; 12:100553. [PMID: 38357385 PMCID: PMC10864763 DOI: 10.1016/j.ejro.2024.100553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/16/2024] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is a common and lethal cancer. From diagnosis to disease staging, response to neoadjuvant therapy assessment and patient surveillance after resection, imaging plays a central role, guiding the multidisciplinary team in decision-planning. Review aims and findings This review discusses the most up-to-date imaging recommendations, typical and atypical findings, and issues related to each step of patient management. Example cases for each relevant condition are presented, and a structured report for disease staging is suggested. Conclusion Despite current issues in PDAC imaging at different stages of patient management, the radiologist is essential in the multidisciplinary team, as the conveyor of relevant imaging findings crucial for patient care.
Collapse
Affiliation(s)
- Carlos Bilreiro
- Radiology Department, Champalimaud Foundation, Lisbon, Portugal
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
- Nova Medical School, Lisbon, Portugal
| | - Luísa Andrade
- Radiology Department, Champalimaud Foundation, Lisbon, Portugal
| | - Inês Santiago
- Radiology Department, Champalimaud Foundation, Lisbon, Portugal
| | - Rui Mateus Marques
- Nova Medical School, Lisbon, Portugal
- Radiology Department, Hospital de S. José, Lisbon, Portugal
| | - Celso Matos
- Radiology Department, Champalimaud Foundation, Lisbon, Portugal
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| |
Collapse
|
11
|
Pathak P, Abandeh L, Aboughalia H, Pooyan A, Mansoori B. Overview of F18-FDG uptake patterns in retroperitoneal pathologies: imaging findings, pitfalls, and artifacts. Abdom Radiol (NY) 2024; 49:1677-1698. [PMID: 38652126 DOI: 10.1007/s00261-023-04139-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/14/2023] [Accepted: 11/22/2023] [Indexed: 04/25/2024]
Abstract
INTRODUCTION Retroperitoneum can be the origin of a wide variety of pathologic conditions and potential space for disease spread to other compartments of the abdomen and pelvis. Computed tomography (CT) and magnetic resonance imaging (MRI) are often the initial imaging modalities to evaluate the retroperitoneal pathologies, however given the intrinsic limitations, F18-FDG PET/CT provides additional valuable metabolic information which can change the patient management and clinical outcomes. We highlight the features of retroperitoneal pathologies on F18-FDG PET/CT and the commonly encountered imaging artifacts and pitfalls. The aim of this review is to characterize primary and secondary retroperitoneal pathologies based on their metabolic features, and correlate PET findings with anatomic imaging. CONCLUSION Retroperitoneal pathologies can be complex, ranging from oncologic to a spectrum of non-oncologic disorders. While crosse-sectional imaging (CT and MRI) are often the initial imaging modalities to localize and characterize pathologies, metabolic information provided by F18-FDG PET/CT can change the management and clinical outcome in many cases.
Collapse
Affiliation(s)
- Priya Pathak
- Abdominal Imaging and Nuclear Medicine Divisions, Department of Radiology, University of Minnesota, 420 Delaware St SE, Minneapolis, MN, 55455, USA.
| | - Laith Abandeh
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Hassan Aboughalia
- Department of Radiology, Children's National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Atefe Pooyan
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Bahar Mansoori
- Abdominal Imaging Division, Department of Radiology, University of Washington, Seattle, WA, USA
| |
Collapse
|
12
|
Bartlett DJ, Takahashi H, Bach CR, Lunn B, Thorpe MP, Broski SM, Packard AT, Fletcher JG, Navin PJ. Potential applications of PET/MRI in non-oncologic conditions within the abdomen and pelvis. Abdom Radiol (NY) 2023; 48:3624-3633. [PMID: 37145312 DOI: 10.1007/s00261-023-03922-0] [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: 02/21/2023] [Revised: 04/03/2023] [Accepted: 04/12/2023] [Indexed: 05/06/2023]
Abstract
PET/MRI is a relatively new imaging modality with several advantages over PET/CT that promise to improve imaging of the abdomen and pelvis for specific diagnostic tasks by combining the superior soft tissue characterization of MRI with the functional information acquired from PET. PET/MRI has an established role in staging and response assessment of multiple abdominopelvic malignancies, but the modality is not yet established for non-oncologic conditions of the abdomen and pelvis. In this review, potential applications of PET/MRI for non-oncologic conditions of abdomen and pelvis are outlined, and the available literature is reviewed to highlight promising areas for further research and translation into clinical practice.
Collapse
Affiliation(s)
| | | | - Corrie R Bach
- Department of Radiology, Mayo Clinic, Rochester, USA
| | - Brendan Lunn
- Department of Radiology, Mayo Clinic, Rochester, USA
| | | | | | - Ann T Packard
- Department of Radiology, Mayo Clinic, Rochester, USA
| | | | | |
Collapse
|
13
|
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
|
14
|
Oba A, Del Chiaro M, Fujii T, Okano K, Stoop TF, Wu YHA, Maekawa A, Yoshida Y, Hashimoto D, Sugawara T, Inoue Y, Tanabe M, Sho M, Sasaki T, Takahashi Y, Matsumoto I, Sasahira N, Nagakawa Y, Satoi S, Schulick RD, Yoon YS, He J, Jang JY, Wolfgang CL, Hackert T, Besselink MG, Takaori K, Takeyama Y. "Conversion surgery" for locally advanced pancreatic cancer: A position paper by the study group at the joint meeting of the International Association of Pancreatology (IAP) & Japan Pancreas Society (JPS) 2022. Pancreatology 2023; 23:712-720. [PMID: 37336669 DOI: 10.1016/j.pan.2023.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 04/10/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023]
Abstract
Locally advanced pancreatic cancer (LAPC), which progresses locally and surrounds major vessels, has historically been deemed unresectable. Surgery alone failed to provide curative resection and improve overall survival. With the advancements in treatment, reports have shown favorable results in LAPC after undergoing successful chemotherapy therapy or chemoradiation therapy followed by surgical resection, so-called "conversion surgery", at experienced high-volume centers. However, recognizing significant regional and institutional disparities in the management of LAPC, an international consensus meeting on conversion surgery for LAPC was held during the Joint Congress of the 26th Meeting of the International Association of Pancreatology (IAP) and the 53rd Annual Meeting of Japan Pancreas Society (JPS) in Kyoto in July 2022. During the meeting, presenters reported the current best multidisciplinary practices for LAPC, including preoperative modalities, best systemic treatment regimens and durations, procedures of conversion surgery with or without vascular resections, biomarkers, and genetic studies. It was unanimously agreed among the experts in this meeting that "cancer biology is surpassing locoregional anatomical resectability" in the era of effective multiagent treatment. The biology of pancreatic cancer has yet to be further elucidated, and we believe it is essential to improve the treatment outcomes of LAPC patients through continued efforts from each institution and more international collaboration. This article summarizes the agreement during the discussion amongst the experts in the meeting. We hope that this will serve as a foundation for future international collaboration and recommendations for future guidelines.
Collapse
Affiliation(s)
- Atsushi Oba
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan; Division of Surgical Oncology, Department of Surgery, University of Colorado of Medicine, Anschutz Medical Campus, Aurora, CO, USA; Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Marco Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado of Medicine, Anschutz Medical Campus, Aurora, CO, USA.
| | - Tsutomu Fujii
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Keiichi Okano
- Department of Gastroenterological Surgery, Kagawa University School of Medicine, Kagawa, Japan
| | - Thomas F Stoop
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Y H Andrew Wu
- Department Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aya Maekawa
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan; Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuta Yoshida
- Department of Surgery, Kindai University, Osaka, Japan
| | | | - Toshitaka Sugawara
- Division of Surgical Oncology, Department of Surgery, University of Colorado of Medicine, Anschutz Medical Campus, Aurora, CO, USA; Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yosuke Inoue
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | - Minoru Tanabe
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masayuki Sho
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Takashi Sasaki
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | - Yu Takahashi
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | | | - Naoki Sasahira
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | - Yuichi Nagakawa
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, Tokyo, Japan
| | - Sohei Satoi
- Division of Surgical Oncology, Department of Surgery, University of Colorado of Medicine, Anschutz Medical Campus, Aurora, CO, USA; Department of Surgery, Kansai Medical University, Osaka, Japan
| | - Richard D Schulick
- Division of Surgical Oncology, Department of Surgery, University of Colorado of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Yoo-Seok Yoon
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jin He
- Department Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | | | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Germany; Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Germany
| | - Marc G Besselink
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | | | | |
Collapse
|
15
|
Jain AJ, Maxwell JE, Katz MHG, Snyder RA. Surgical Considerations for Neoadjuvant Therapy for Pancreatic Adenocarcinoma. Cancers (Basel) 2023; 15:4174. [PMID: 37627202 PMCID: PMC10453019 DOI: 10.3390/cancers15164174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a challenging disease process with a 5-year survival rate of only 11%. Neoadjuvant therapy in patients with localized pancreatic cancer has multiple theoretical benefits, including improved patient selection for surgery, early delivery of systemic therapy, and assessment of response to therapy. Herein, we review key surgical considerations when selecting patients for neoadjuvant therapy and curative-intent resection. Accurate determination of resectability at diagnosis is critical and should be based on not only anatomic criteria but also biologic and clinical criteria to determine optimal treatment sequencing. Borderline resectable or locally advanced pancreatic cancer is best treated with neoadjuvant therapy and resection, including vascular resection and reconstruction when appropriate. Lastly, providing nutritional, prehabilitation, and supportive care interventions to improve patient fitness prior to surgical intervention and adequately address the adverse effects of therapy is critical.
Collapse
Affiliation(s)
| | | | | | - Rebecca A. Snyder
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.J.)
| |
Collapse
|
16
|
Jan IS, Ch'ang HJ. Selection of patients with pancreatic adenocarcinoma who may benefit from radiotherapy. Radiat Oncol 2023; 18:137. [PMID: 37596627 PMCID: PMC10439654 DOI: 10.1186/s13014-023-02328-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 08/03/2023] [Indexed: 08/20/2023] Open
Abstract
Despite combination chemotherapy demonstrating a positive effect on survival, the clinical outcomes of pancreatic adenocarcinoma (PDAC) remain poor. Radiotherapy was previously a component of the curative treatment of PDAC. Advances in imaging and computer sciences have enabled the prescription of higher dosage of radiation focused on tumours with minimal toxicity to normal tissue. However, the role of radiotherapy has not been established in the curative treatment of localized PDAC because of the conflicting results from large prospective trials. Most studies have demonstrated improved locoregional control but no survival benefit from additional chemoradiotherapy (CRT) in addition to chemotherapy for resectable, borderline or locally advanced PDAC. The improved locoregional control enabled by CRT does not cause extended survival because of rapid distant progression in a significant proportion of patients with PDAC. Several single-institute studies of prescribing intensive chemotherapy with modern ablative radiotherapy for locally advanced PDAC have demonstrated extended survival with an acceptable safety profile. In an analysis after long-term follow-up, the PREOPANC study demonstrated a survival benefit from neoadjuvant gemcitabine-based CRT in resected PDAC relative to upfront surgery followed by adjuvant gemcitabine only. These observations indicated that the role of radiotherapy in PDAC should be evaluated in a subgroup of patients without rapid distant progression because systemic therapy for PDAC remains underdeveloped. We reviewed critical imaging, tissue, liquid and clinical biomarkers to differentiate the heterogeneous biologic spectra of patients with PDAC to identify those who may benefit the most from local radiotherapy. Exclusion of patients with localised PDAC who develop distant progression in a short time and undergo extended upfront chemotherapy for over 4 months may enable the identification of a survival benefit of local radiotherapy. Though promising, the effectiveness of biomarkers must be validated in a multi-institutional prospective study of patients with PDAC receiving CRT or not receiving CRT.
Collapse
Affiliation(s)
- I-Shiow Jan
- Department of Laboratory Medicine, College of Medicine, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
| | - Hui Ju Ch'ang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan.
- Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
- Department of Radiation Oncology, Taipei Medical University, Taipei, Taiwan.
- Department of Oncology, School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
17
|
Mukherjee S, Korfiatis P, Khasawneh H, Rajamohan N, Patra A, Suman G, Singh A, Thakkar J, Patnam NG, Trivedi KH, Karbhari A, Chari ST, Truty MJ, Halfdanarson TR, Bolan CW, Sandrasegaran K, Majumder S, Goenka AH. Bounding box-based 3D AI model for user-guided volumetric segmentation of pancreatic ductal adenocarcinoma on standard-of-care CTs. Pancreatology 2023; 23:522-529. [PMID: 37296006 PMCID: PMC10676442 DOI: 10.1016/j.pan.2023.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023]
Abstract
OBJECTIVES To develop a bounding-box-based 3D convolutional neural network (CNN) for user-guided volumetric pancreas ductal adenocarcinoma (PDA) segmentation. METHODS Reference segmentations were obtained on CTs (2006-2020) of treatment-naïve PDA. Images were algorithmically cropped using a tumor-centered bounding box for training a 3D nnUNet-based-CNN. Three radiologists independently segmented tumors on test subset, which were combined with reference segmentations using STAPLE to derive composite segmentations. Generalizability was evaluated on Cancer Imaging Archive (TCIA) (n = 41) and Medical Segmentation Decathlon (MSD) (n = 152) datasets. RESULTS Total 1151 patients [667 males; age:65.3 ± 10.2 years; T1:34, T2:477, T3:237, T4:403; mean (range) tumor diameter:4.34 (1.1-12.6)-cm] were randomly divided between training/validation (n = 921) and test subsets (n = 230; 75% from other institutions). Model had a high DSC (mean ± SD) against reference segmentations (0.84 ± 0.06), which was comparable to its DSC against composite segmentations (0.84 ± 0.11, p = 0.52). Model-predicted versus reference tumor volumes were comparable (mean ± SD) (29.1 ± 42.2-cc versus 27.1 ± 32.9-cc, p = 0.69, CCC = 0.93). Inter-reader variability was high (mean DSC 0.69 ± 0.16), especially for smaller and isodense tumors. Conversely, model's high performance was comparable between tumor stages, volumes and densities (p > 0.05). Model was resilient to different tumor locations, status of pancreatic/biliary ducts, pancreatic atrophy, CT vendors and slice thicknesses, as well as to the epicenter and dimensions of the bounding-box (p > 0.05). Performance was generalizable on MSD (DSC:0.82 ± 0.06) and TCIA datasets (DSC:0.84 ± 0.08). CONCLUSION A computationally efficient bounding box-based AI model developed on a large and diverse dataset shows high accuracy, generalizability, and robustness to clinically encountered variations for user-guided volumetric PDA segmentation including for small and isodense tumors. CLINICAL RELEVANCE AI-driven bounding box-based user-guided PDA segmentation offers a discovery tool for image-based multi-omics models for applications such as risk-stratification, treatment response assessment, and prognostication, which are urgently needed to customize treatment strategies to the unique biological profile of each patient's tumor.
Collapse
Affiliation(s)
- Sovanlal Mukherjee
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Panagiotis Korfiatis
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Hala Khasawneh
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Naveen Rajamohan
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Anurima Patra
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Garima Suman
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Aparna Singh
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Jay Thakkar
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Nandakumar G Patnam
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Kamaxi H Trivedi
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Aashna Karbhari
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Suresh T Chari
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Mark J Truty
- Department of Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | | | - Candice W Bolan
- Department of Radiology, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL, 32224, USA.
| | - Kumar Sandrasegaran
- Department of Radiology, Mayo Clinic, 13400 E Shea Blvd, Scottsdale, AZ, 85259, USA.
| | - Shounak Majumder
- Department of Gastroenterology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Ajit H Goenka
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
18
|
Tu W, Gottumukkala RV, Schieda N, Lavallée L, Adam BA, Silverman SG. Perineural Invasion and Spread in Common Abdominopelvic Diseases: Imaging Diagnosis and Clinical Significance. Radiographics 2023; 43:e220148. [PMID: 37319024 DOI: 10.1148/rg.220148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Malignancies and other diseases may spread by multiple pathways, including direct extension, hematogenous spread, or via lymphatic vessels. A less-well-understood route is the peripheral nervous system, which is known as perineural spread (PNS). In addition to accounting for pain and other neurologic symptoms, PNS affects both disease prognosis and management. Although PNS is commonly discussed in relation to head and neck tumors, there is emerging data regarding PNS in abdominopelvic malignancies and other conditions such as endometriosis. Due to improved contrast and spatial resolution, perineural invasion, a finding heretofore diagnosed only at pathologic examination, can be detected at CT, MRI, and PET/CT. PNS most commonly manifests as abnormal soft-tissue attenuation extending along neural structures, and diagnosis of it is aided by optimizing imaging parameters, understanding pertinent anatomy, and becoming familiar with the typical neural pathways of spread that largely depend on the disease type and location. In the abdomen, the celiac plexus is a central structure that innervates the major abdominal organs and is the principal route of PNS in patients with pancreatic and biliary carcinomas. In the pelvis, the lumbosacral plexus and inferior hypogastric plexus are the central structures and principal routes of PNS in patients with pelvic malignancies. Although the imaging findings of PNS may be subtle, a radiologic diagnosis can have a substantial effect on patient care. Knowledge of anatomy and known routes of PNS and optimizing imaging parameters is of utmost importance in providing key information for prognosis and treatment planning. © RSNA, 2023 Supplemental material and the slide presentation from the RSNA Annual Meeting are available for this article. Quiz questions for this article are available through the Online Learning Center.
Collapse
Affiliation(s)
- Wendy Tu
- From the Department of Radiology and Diagnostic Imaging (W.T.) and Department of Laboratory Medicine and Pathology (B.A.A.), University of Alberta, 116 St & 85 Ave, Edmonton, AB, Canada T6G 2R3; Department of Radiology, Brigham and Women's Hospital, Harvard University, Boston, Mass (R.V.G., S.G.S.); and Departments of Radiology (N.S.) and Urology (L.L.), University of Ottawa, Ottawa, Ontario, Canada
| | - Ravi V Gottumukkala
- From the Department of Radiology and Diagnostic Imaging (W.T.) and Department of Laboratory Medicine and Pathology (B.A.A.), University of Alberta, 116 St & 85 Ave, Edmonton, AB, Canada T6G 2R3; Department of Radiology, Brigham and Women's Hospital, Harvard University, Boston, Mass (R.V.G., S.G.S.); and Departments of Radiology (N.S.) and Urology (L.L.), University of Ottawa, Ottawa, Ontario, Canada
| | - Nicola Schieda
- From the Department of Radiology and Diagnostic Imaging (W.T.) and Department of Laboratory Medicine and Pathology (B.A.A.), University of Alberta, 116 St & 85 Ave, Edmonton, AB, Canada T6G 2R3; Department of Radiology, Brigham and Women's Hospital, Harvard University, Boston, Mass (R.V.G., S.G.S.); and Departments of Radiology (N.S.) and Urology (L.L.), University of Ottawa, Ottawa, Ontario, Canada
| | - Luke Lavallée
- From the Department of Radiology and Diagnostic Imaging (W.T.) and Department of Laboratory Medicine and Pathology (B.A.A.), University of Alberta, 116 St & 85 Ave, Edmonton, AB, Canada T6G 2R3; Department of Radiology, Brigham and Women's Hospital, Harvard University, Boston, Mass (R.V.G., S.G.S.); and Departments of Radiology (N.S.) and Urology (L.L.), University of Ottawa, Ottawa, Ontario, Canada
| | - Benjamin A Adam
- From the Department of Radiology and Diagnostic Imaging (W.T.) and Department of Laboratory Medicine and Pathology (B.A.A.), University of Alberta, 116 St & 85 Ave, Edmonton, AB, Canada T6G 2R3; Department of Radiology, Brigham and Women's Hospital, Harvard University, Boston, Mass (R.V.G., S.G.S.); and Departments of Radiology (N.S.) and Urology (L.L.), University of Ottawa, Ottawa, Ontario, Canada
| | - Stuart G Silverman
- From the Department of Radiology and Diagnostic Imaging (W.T.) and Department of Laboratory Medicine and Pathology (B.A.A.), University of Alberta, 116 St & 85 Ave, Edmonton, AB, Canada T6G 2R3; Department of Radiology, Brigham and Women's Hospital, Harvard University, Boston, Mass (R.V.G., S.G.S.); and Departments of Radiology (N.S.) and Urology (L.L.), University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
19
|
Yoo J, Lee JM, Joo I, Lee DH, Yoon JH, Yu MH, Jang JY, Lee SH. Post-neoadjuvant treatment pancreatic cancer resectability and outcome prediction using CT, 18F-FDG PET/MRI and CA 19-9. Cancer Imaging 2023; 23:49. [PMID: 37217958 DOI: 10.1186/s40644-023-00565-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/01/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND CT prediction of resectability and prognosis following neoadjuvant treatment (NAT) in patients with pancreatic ductal adenocarcinoma (PDAC) remains challenging. This study aims to determine whether addition of 18F-fluorodeoxyglucose (FDG) postiron emission tomography (PET)/MRI and carbohydrate antigen (CA) 19-9 to contrast-enhanced CT (CECT) can improve accuracy of predicting resectability compared to CECT alone and predict prognosis in PDAC patients after NAT. METHODS In this retrospective study, 120 PDAC patients (65 women; mean age, 66.7 years [standard deviation, 8.4]) underwent CECT, PET/MRI, and CA 19-9 examinations after NAT between January 2013 and June 2021. Three board-certified radiologists independently rated the overall resectability on a 5-point scale (score 5, definitely resectable) in three sessions (session 1, CECT; 2, CECT plus PET/MRI─no FDG avidity and no diffusion restriction at tumor-vessel contact indicated modification of CECT scores to ≥ 3; 3, CECT plus PET plus CA 19-9─no FDG avidity at tumor-vessel contact and normalized CA 19-9 indicated modification of CECT scores to ≥ 3). Jackknife free-response receiver operating characteristic method and generalized estimating equations were used to compare pooled area under the curve (AUC), sensitivity, and specificity of three sessions. Predictors for recurrence-free survival (RFS) were assessed using Cox regression analyses. RESULTS Each session showed different pooled AUC (session 1 vs. 2 vs. 3, 0.853 vs. 0.873 vs. 0.874, p = 0.026), sensitivity (66.2% [137/207] vs. 86.0% [178/207] vs. 84.5% [175/207], p < 0.001) and specificity (67.3% [103/153] vs. 58.8% [90/153] vs. 60.1% [92/153], p = 0.048). According to pairwise comparison, specificity of CECT plus PET/MRI was lower than that of CECT alone (adjusted p = 0.042), while there was no significant difference in specificity between CECT alone and CECT plus PET plus CA 19-9 (adjusted p = 0.081). Twenty-eight of 69 patients (40.6%) with R0 resection experienced tumor recurrence (mean follow-up, 18.0 months). FDG avidity at tumor-vessel contact on post-NAT PET (HR = 4.37, p = 0.033) and pathologically confirmed vascular invasion (HR = 5.36, p = 0.004) predicted RFS. CONCLUSION Combination of CECT, PET and CA 19-9 increased area under the curve and sensitivity for determining resectability, compared to CECT alone, without compromising the specificity. Furthermore, 18F-FDG avidity at tumor-vessel contact on post-NAT PET predicted RFS.
Collapse
Affiliation(s)
- Jeongin Yoo
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Jeong Min Lee
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.
- Department of Radiology, Seoul National University College of Medicine, Seoul, Korea.
- Institute of Radiation Medicine, Seoul National University College of Medicine, Seoul, Korea.
- Department of Radiology, Konkuk University School of Medicine, Seoul, Korea.
| | - Ijin Joo
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
- Institute of Radiation Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Radiology, Konkuk University School of Medicine, Seoul, Korea
| | - Dong Ho Lee
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong Hee Yoon
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Mi Hye Yu
- Institute of Radiation Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Radiology, Konkuk University School of Medicine, Seoul, Korea
| | - Jin-Young Jang
- Department of General Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Hyub Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
20
|
Guggenberger KV, Bley TA, Held S, Keller R, Flemming S, Wiegering A, Germer CT, Kimmel B, Kunzmann V, Hartlapp I, Anger F. Predictive value of computed tomography on surgical resectability in locally advanced pancreatic cancer treated with multiagent induction chemotherapy: Results from a prospective, multicentre phase 2 trial (NEOLAP-AIO-PAK-0113). Eur J Radiol 2023; 163:110834. [PMID: 37080059 DOI: 10.1016/j.ejrad.2023.110834] [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: 03/06/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/22/2023]
Abstract
PURPOSE To assess the role of current imaging-based resectability criteria and the degree of radiological downsizing in locally advanced pancreatic adenocarcinoma (LAPC) after multiagent induction chemotherapy (ICT) in multicentre, open-label, randomized phase 2 trial. METHOD LAPC patients were prospectively treated with multiagent ICT followed by surgical exploration within the NEOLAP trial. All patients underwent CT scan at baseline and after ICT to assess resectability status according to national comprehensive cancer network guidelines (NCCN) criteria and response evaluation criteria in solid tumors (RECIST) at the local study center and retrospectively in a central review. Imaging results were compared in terms of local and central staging, downsizing and pathological resection status. RESULTS 83 patients were evaluable for central review of baseline and restaging imaging results. Downstaging by central review was rarely seen after multiagent ICT (7.7%), whereas tumor downsizing was documented frequently (any downsizing 90.4%, downsizing to partial response (PR) according to RECIST: 26.5%). Patients with any downsizing showed no significant different R0 resection rate (37.3%) as patients that fulfilled the criteria of PR (40.9%). The sensitivity of any downsizing for predicting R0 resection was 97% with a negative predictive value (NPV) of 0.88. ROC-analysis revealed that tumor downsizing was a predictor of R0 resection (AUC 0.647, p = 0.028) with a best cut-off value of 22.5% downsizing yielding a sensitivity of 65% and a specificity of 61%. CONCLUSIONS Imaging-based tumor downsizing and not downstaging can guide the selection of patients with a realistic chance of R0-resection in LAPC after multi-agent ICT.
Collapse
Affiliation(s)
- K V Guggenberger
- Department of Diagnostic and Interventional Radiology, University Hospital Wuerzburg, Wuerzburg, Germany.
| | - T A Bley
- Department of Diagnostic and Interventional Radiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - S Held
- Department of Biometrics, ClinAssess GmbH, Leverkusen, Germany
| | - R Keller
- Clinical Research, AIO Studien gGmbH, Berlin, Germany
| | - S Flemming
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - A Wiegering
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - C T Germer
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - B Kimmel
- Department of Internal Medicine II, Medical Oncology and Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
| | - V Kunzmann
- Department of Internal Medicine II, Medical Oncology and Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
| | - I Hartlapp
- Department of Internal Medicine II, Medical Oncology and Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
| | - F Anger
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
21
|
de Jong TL, Koopman D, van der Worp CAJ, Stevens H, Vuijk FA, Vahrmeijer AL, Mieog JSD, de Groot JWB, Meijssen MAC, Nieuwenhuijs VB, de Geus-Oei LF, Jager PL, Patijn GA. Added value of digital FDG-PET/CT in disease staging and restaging in patients with resectable or borderline resectable pancreatic cancer. Surg Oncol 2023; 47:101909. [PMID: 36739788 DOI: 10.1016/j.suronc.2023.101909] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/09/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND We studied the added value of digital FDG-PET/CT in disease staging and restaging compared to the standard work-up with contrast enhanced CT (ceCT) and CA19-9 in patients with resectable or borderline resectable pancreatic cancer who received neo-adjuvant therapy. Primary endpoints were tumor response compared to ceCT and CA19.9 as well as the ability to detect distant metastatic disease. METHODS 35 patients were included in this dual-center prospective study. FDG-PET using digital photon counting technology combined with CT scans were acquired before (T1) and after neo-adjuvant therapy (T2). Patients were staged and restaged based on standard protocol with ceCT and CA 19.9, while all PET/CT scans were stored securely and not included in clinical decision making. After the pancreatic resection, an expert team retrospectively assessed the CT tumor diameter, CA19-9, tumor FDG-uptake, and appearance of metastatic disease of all patients for both time points. RESULTS CA19-9 levels, CT tumor diameter, and tumor FDG-uptake on PET significantly decreased from T1 to T2 (p = 0.017, p = 0.001, and p < 0.0001). The change in FDG-uptake values showed a strong positive correlation with the change in CT tumor diameter and change in CA19-9 (R = 0.75 and R = 0.73, respectively). In addition, small-volume liver lesions were detected on digital PET/CT in 5/35 patients (14%), 4 of which were pathology confirmed at laparotomy. Only one of these five cases was detected on baseline staging ceCT (3%). CONCLUSION We found that adding digital PET/CT strengthens restaging after neo-adjuvant therapy based on the observed strong correlation with ceCT tumor diameter and Ca19.9. Also, digital PET/CT was found to detect occult metastatic disease not visualized on ceCT, that would have resulted in altered disease staging and therapeutic strategy in a substantial proportion of patients.
Collapse
Affiliation(s)
- Tonke L de Jong
- Department of Nuclear Medicine, Isala Hospital, Zwolle, the Netherlands
| | - Daniëlle Koopman
- Department of Nuclear Medicine, Isala Hospital, Zwolle, the Netherlands
| | | | - Henk Stevens
- Department of Nuclear Medicine, Isala Hospital, Zwolle, the Netherlands
| | - Floris A Vuijk
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Maarten A C Meijssen
- Department of Gastroenterology and Hepatology, Isala Hospital, Zwolle, the Netherlands
| | | | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Biomedical Photonic Imaging Group, University of Twente, Enschede, the Netherlands
| | - Pieter L Jager
- Department of Nuclear Medicine, Isala Hospital, Zwolle, the Netherlands
| | - Gijs A Patijn
- Department of Surgery, Isala Hospital, Zwolle, the Netherlands.
| |
Collapse
|
22
|
Furtado FS, Mercaldo ND, Vahle T, Benkert T, Bradley WR, Ratanaprasatporn L, Seethamraju RT, Harisinghani MG, Lee S, Suarez-Weiss K, Umutlu L, Catana C, Pomykala KL, Domachevsky L, Bernstine H, Groshar D, Rosen BR, Catalano OA. Simultaneous multislice diffusion-weighted imaging versus standard diffusion-weighted imaging in whole-body PET/MRI. Eur Radiol 2023; 33:2536-2547. [PMID: 36460925 DOI: 10.1007/s00330-022-09275-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/20/2022] [Accepted: 10/26/2022] [Indexed: 12/04/2022]
Abstract
OBJECTIVE To compare standard (STD-DWI) single-shot echo-planar imaging DWI and simultaneous multislice (SMS) DWI during whole-body positron emission tomography (PET)/MRI regarding acquisition time, image quality, and lesion detection. METHODS Eighty-three adults (47 females, 57%), median age of 64 years (IQR 52-71), were prospectively enrolled from August 2018 to March 2020. Inclusion criteria were (a) abdominal or pelvic tumors and (b) PET/MRI referral from a clinician. Patients were excluded if whole-body acquisition of STD-DWI and SMS-DWI sequences was not completed. The evaluated sequences were axial STD-DWI at b-values 50-400-800 s/mm2 and the apparent diffusion coefficient (ADC), and axial SMS-DWI at b-values 50-300-800 s/mm2 and ADC, acquired with a 3-T PET/MRI scanner. Three radiologists rated each sequence's quality on a five-point scale. Lesion detection was quantified using the anatomic MRI sequences and PET as the reference standard. Regression models were constructed to quantify the association between all imaging outcomes/scores and sequence type. RESULTS The median whole-body STD-DWI acquisition time was 14.8 min (IQR 14.1-16.0) versus 7.0 min (IQR 6.7-7.2) for whole-body SMS-DWI, p < 0.001. SMS-DWI image quality scores were higher than STD-DWI in the abdomen (OR 5.31, 95% CI 2.76-10.22, p < 0.001), but lower in the cervicothoracic junction (OR 0.21, 95% CI 0.10-0.43, p < 0.001). There was no significant difference in the chest, mediastinum, pelvis, and rectum. STD-DWI detected 276/352 (78%) lesions while SMS-DWI located 296/352 (84%, OR 1.46, 95% CI 1.02-2.07, p = 0.038). CONCLUSIONS In cancer staging and restaging, SMS-DWI abbreviates acquisition while maintaining or improving the diagnostic yield in most anatomic regions. KEY POINTS • Simultaneous multislice diffusion-weighted imaging enables faster whole-body image acquisition. • Simultaneous multislice diffusion-weighted imaging maintains or improves image quality when compared to single-shot echo-planar diffusion-weighted imaging in most anatomical regions. • Simultaneous multislice diffusion-weighted imaging leads to superior lesion detection.
Collapse
Affiliation(s)
- Felipe S Furtado
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School, 149 13th Street, Charlestown, MA, 02129, USA
| | - Nathaniel D Mercaldo
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Thomas Vahle
- MR Application Predevelopment, Siemens Healthcare GmbH, Allee am Roethelheimpark 2, 91052, Erlangen, Germany
| | - Thomas Benkert
- MR Application Predevelopment, Siemens Healthcare GmbH, Allee am Roethelheimpark 2, 91052, Erlangen, Germany
| | - William R Bradley
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Lisa Ratanaprasatporn
- Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA
| | - Ravi Teja Seethamraju
- Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School, 149 13th Street, Charlestown, MA, 02129, USA
- MR Collaborations, Siemens Medical Solutions USA, Inc., 30 Jonathan Ln, Malden, MA, 02148, USA
| | - Mukesh G Harisinghani
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Susanna Lee
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Krista Suarez-Weiss
- Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA
| | - Lale Umutlu
- Universitätsmedizin Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Ciprian Catana
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School, 149 13th Street, Charlestown, MA, 02129, USA
| | | | - Liran Domachevsky
- Sheba Medical Center, Derech Sheba 2, Ramat Gan, Israel
- Tel Aviv University, 6997801, Tel Aviv-Yafo, Israel
| | - Hanna Bernstine
- Tel Aviv University, 6997801, Tel Aviv-Yafo, Israel
- Assuta Medical Center, HaBarzel 20 St, Ramat Hahayal, Tel Aviv, Israel
| | - David Groshar
- Tel Aviv University, 6997801, Tel Aviv-Yafo, Israel
- Assuta Medical Center, HaBarzel 20 St, Ramat Hahayal, Tel Aviv, Israel
| | - Bruse R Rosen
- Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School, 149 13th Street, Charlestown, MA, 02129, USA
| | - Onofrio Antonio Catalano
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School, 149 13th Street, Charlestown, MA, 02129, USA.
| |
Collapse
|
23
|
Clinical Implications of FDG-PET in Pancreatic Ductal Adenocarcinoma Patients Treated with Neoadjuvant Therapy. J Gastrointest Surg 2023; 27:337-346. [PMID: 36652179 DOI: 10.1007/s11605-023-05591-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/07/2023] [Indexed: 01/19/2023]
Abstract
PURPOSE To evaluate the clinical significance of 18F-fluorodeoxyglucose positron emission tomography/computed tomography in patients with pancreatic ductal adenocarcinoma who underwent neoadjuvant therapy. METHODS Among 285 consecutive patients who underwent pancreatic resection for pancreatic ductal adenocarcinoma between 2015 and 2021, 86 who underwent preoperative 18F-fluorodeoxyglucose positron emission tomography/computed tomography after completion of neoadjuvant treatment were reviewed. Among preoperative factors, including post-treatment maximum standardized uptake value, predictors of early recurrence and poor prognosis were identified using multivariate analysis for decision making in surgery. RESULTS Nineteen (22%) patients with pancreatic ductal adenocarcinoma demonstrated high maximum standardized uptake (≥ 4.5). High post-treatment maximum standardized uptake (≥ 4.5) predicted early recurrence within 6 months after surgery and correlated with shorter recurrence-free survival. Elevated post-treatment CA19-9 level (> 37 U/ml) and maximum standardized uptake ≥ 4.5 were independent prognostic factors. Post-treatment, a high maximum standardized uptake value indicated a poorer prognosis than a low maximum standardized uptake value in both patients with elevated CA19-9 and normal CA19-9 levels. The median overall survival in patients with elevated post-treatment CA19-9 and high maximum standardized uptake was only 17 months; 67% experienced early recurrence. Dynamic changes in maximum standardized uptake during neoadjuvant therapy were correlated with pathological response to neoadjuvant therapy, but not with radiological response or change in CA19-9 level. CONCLUSIONS Post-treatment assessment using maximum standardized uptake value is useful for stratifying patients with pancreatic ductal adenocarcinoma who will benefit from surgery. Instead of subsequent curative resection, additional neoadjuvant therapy should be considered in patients with a persistently high maximum standardized uptake value.
Collapse
|
24
|
Li J, Fu C, Zhao S, Pu Y, Yang F, Zeng S, Yang C, Gao H, Chen L. The progress of PET/MRI in clinical management of patients with pancreatic malignant lesions. Front Oncol 2023; 13:920896. [PMID: 37188192 PMCID: PMC10175752 DOI: 10.3389/fonc.2023.920896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Recently, the morbidity and mortality of pancreatic cancer have been increasing year by year. Because of its deep anatomical location and because most presented patients often suffer from abdominal pain or jaundice, it is difficult to diagnose pancreatic cancer at an early stage, leading to late clinical stage and poor prognosis. integrated positron emission tomography/magnetic resonance imaging (PET/MRI) fusion imaging not only has the characteristics of high resolution and multi-parameter imaging of MRI, but also combines the high sensitivity and the semi-quantitative characteristics of PET. In addition, the continuous development of novel MRI imaging and PET imaging biomarkers provide a unique and precise research direction for future pancreatic cancer research. This review summarizes the value of PET/MRI in the diagnosis, staging, efficacy monitoring, and prognosis evaluation of pancreatic cancer, and prognosis for developing emerging imaging agents and artificial intelligence radiomics in pancreatic cancer.
Collapse
Affiliation(s)
- Jindan Li
- Department of PET-CT/MR Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Chaojiang Fu
- Department of Emergency, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Sheng Zhao
- Department of PET-CT/MR Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yongzhu Pu
- Department of PET-CT/MR Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Fake Yang
- Department of PET-CT/MR Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Shuguang Zeng
- Department of Information Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Conghui Yang
- Department of PET-CT/MR Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hongqiang Gao
- Department of Hepatobiliary Surgery, The First People’s Hospital of Kunming City & Ganmei Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Long Chen, ; Hongqiang Gao,
| | - Long Chen
- Department of PET-CT/MR Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
- *Correspondence: Long Chen, ; Hongqiang Gao,
| |
Collapse
|
25
|
Mukherjee S, Patra A, Khasawneh H, Korfiatis P, Rajamohan N, Suman G, Majumder S, Panda A, Johnson MP, Larson NB, Wright DE, Kline TL, Fletcher JG, Chari ST, Goenka AH. Radiomics-based Machine-learning Models Can Detect Pancreatic Cancer on Prediagnostic Computed Tomography Scans at a Substantial Lead Time Before Clinical Diagnosis. Gastroenterology 2022; 163:1435-1446.e3. [PMID: 35788343 DOI: 10.1053/j.gastro.2022.06.066] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND & AIMS Our purpose was to detect pancreatic ductal adenocarcinoma (PDAC) at the prediagnostic stage (3-36 months before clinical diagnosis) using radiomics-based machine-learning (ML) models, and to compare performance against radiologists in a case-control study. METHODS Volumetric pancreas segmentation was performed on prediagnostic computed tomography scans (CTs) (median interval between CT and PDAC diagnosis: 398 days) of 155 patients and an age-matched cohort of 265 subjects with normal pancreas. A total of 88 first-order and gray-level radiomic features were extracted and 34 features were selected through the least absolute shrinkage and selection operator-based feature selection method. The dataset was randomly divided into training (292 CTs: 110 prediagnostic and 182 controls) and test subsets (128 CTs: 45 prediagnostic and 83 controls). Four ML classifiers, k-nearest neighbor (KNN), support vector machine (SVM), random forest (RM), and extreme gradient boosting (XGBoost), were evaluated. Specificity of model with highest accuracy was further validated on an independent internal dataset (n = 176) and the public National Institutes of Health dataset (n = 80). Two radiologists (R4 and R5) independently evaluated the pancreas on a 5-point diagnostic scale. RESULTS Median (range) time between prediagnostic CTs of the test subset and PDAC diagnosis was 386 (97-1092) days. SVM had the highest sensitivity (mean; 95% confidence interval) (95.5; 85.5-100.0), specificity (90.3; 84.3-91.5), F1-score (89.5; 82.3-91.7), area under the curve (AUC) (0.98; 0.94-0.98), and accuracy (92.2%; 86.7-93.7) for classification of CTs into prediagnostic versus normal. All 3 other ML models, KNN, RF, and XGBoost, had comparable AUCs (0.95, 0.95, and 0.96, respectively). The high specificity of SVM was generalizable to both the independent internal (92.6%) and the National Institutes of Health dataset (96.2%). In contrast, interreader radiologist agreement was only fair (Cohen's kappa 0.3) and their mean AUC (0.66; 0.46-0.86) was lower than each of the 4 ML models (AUCs: 0.95-0.98) (P < .001). Radiologists also recorded false positive indirect findings of PDAC in control subjects (n = 83) (7% R4, 18% R5). CONCLUSIONS Radiomics-based ML models can detect PDAC from normal pancreas when it is beyond human interrogation capability at a substantial lead time before clinical diagnosis. Prospective validation and integration of such models with complementary fluid-based biomarkers has the potential for PDAC detection at a stage when surgical cure is a possibility.
Collapse
Affiliation(s)
| | - Anurima Patra
- Department of Radiology, Tata Medical Centre, Kolkata, India
| | - Hala Khasawneh
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Garima Suman
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Shounak Majumder
- Department of Gastroenterology, Mayo Clinic, Rochester, Minnesota
| | - Ananya Panda
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Matthew P Johnson
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Nicholas B Larson
- Department of Radiology, Mayo Clinic, Rochester, Minnesota; Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | | | | | | | - Suresh T Chari
- Department of Gastroenterology, Mayo Clinic, Rochester, Minnesota; Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ajit H Goenka
- Department of Radiology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
26
|
Beyond the AJR: FDG PET/MRI Has the Potential to Improve the Detection of Peritoneal Carcinomatosis Compared With Standard-of-Care Imaging. AJR Am J Roentgenol 2022; 219:845. [PMID: 35383488 DOI: 10.2214/ajr.22.27750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
27
|
Wright DE, Mukherjee S, Patra A, Khasawneh H, Korfiatis P, Suman G, Chari ST, Kudva YC, Kline TL, Goenka AH. Radiomics-based machine learning (ML) classifier for detection of type 2 diabetes on standard-of-care abdomen CTs: a proof-of-concept study. Abdom Radiol (NY) 2022; 47:3806-3816. [PMID: 36085379 DOI: 10.1007/s00261-022-03668-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 01/18/2023]
Abstract
PURPOSE To determine if pancreas radiomics-based AI model can detect the CT imaging signature of type 2 diabetes (T2D). METHODS Total 107 radiomic features were extracted from volumetrically segmented normal pancreas in 422 T2D patients and 456 age-matched controls. Dataset was randomly split into training (300 T2D, 300 control CTs) and test subsets (122 T2D, 156 control CTs). An XGBoost model trained on 10 features selected through top-K-based selection method and optimized through threefold cross-validation on training subset was evaluated on test subset. RESULTS Model correctly classified 73 (60%) T2D patients and 96 (62%) controls yielding F1-score, sensitivity, specificity, precision, and AUC of 0.57, 0.62, 0.61, 0.55, and 0.65, respectively. Model's performance was equivalent across gender, CT slice thicknesses, and CT vendors (p values > 0.05). There was no difference between correctly classified versus misclassified patients in the mean (range) T2D duration [4.5 (0-15.4) versus 4.8 (0-15.7) years, p = 0.8], antidiabetic treatment [insulin (22% versus 18%), oral antidiabetics (10% versus 18%), both (41% versus 39%) (p > 0.05)], and treatment duration [5.4 (0-15) versus 5 (0-13) years, p = 0.4]. CONCLUSION Pancreas radiomics-based AI model can detect the imaging signature of T2D. Further refinement and validation are needed to evaluate its potential for opportunistic T2D detection on millions of CTs that are performed annually.
Collapse
Affiliation(s)
- Darryl E Wright
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Sovanlal Mukherjee
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Anurima Patra
- Department of Radiology, Tata Medical Center, Kolkata, 700160, India
| | - Hala Khasawneh
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Panagiotis Korfiatis
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Garima Suman
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Suresh T Chari
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
- Department of Gastroenterology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Yogish C Kudva
- Department of Endocrinology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Timothy L Kline
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Ajit H Goenka
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA.
| |
Collapse
|
28
|
Abdelrahman AM, Goenka AH, Alva-Ruiz R, Yonkus JA, Leiting JL, Graham RP, Merrell KW, Thiels CA, Hallemeier CL, Warner SG, Haddock MG, Grotz TE, Tran NH, Smoot RL, Ma WW, Cleary SP, McWilliams RR, Nagorney DM, Halfdanarson TR, Kendrick ML, Truty MJ. FDG-PET Predicts Neoadjuvant Therapy Response and Survival in Borderline Resectable/Locally Advanced Pancreatic Adenocarcinoma. J Natl Compr Canc Netw 2022; 20:1023-1032.e3. [PMID: 36075389 PMCID: PMC12001712 DOI: 10.6004/jnccn.2022.7041] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/03/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Neoadjuvant therapy (NAT) is used in borderline resectable/locally advanced (BR/LA) pancreatic ductal adenocarcinoma (PDAC). Anatomic imaging (CT/MRI) poorly predicts response, and biochemical (CA 19-9) markers are not useful (nonsecretors/nonelevated) in many patients. Pathologic response highly predicts survival post-NAT, but is only known postoperatively. Because metabolic imaging (FDG-PET) reveals primary tumor viability, this study aimed to evaluate our experience with preoperative FDG-PET in patients with BR/LA PDAC in predicting NAT response and survival. METHODS We reviewed all patients with resected BR/LA PDAC who underwent NAT with FDG-PET within 60 days of resection. Pre- and post-NAT metabolic (FDG-PET) and biochemical (CA 19-9) responses were dichotomized in addition to pathologic responses. We compared post-NAT metabolic and biochemical responses as preoperative predictors of pathologic responses and recurrence-free survival (RFS) and overall survival (OS). RESULTS We identified 202 eligible patients. Post-NAT, 58% of patients had optimization of CA 19-9 levels. Major metabolic and pathologic responses were present in 51% and 38% of patients, respectively. Median RFS and OS times were 21 and 48.7 months, respectively. Metabolic response was superior to biochemical response in predicting pathologic response (area under the curve, 0.86 vs 0.75; P<.001). Metabolic response was the only univariate preoperative predictor of OS (odds ratio, 0.25; 95% CI, 0.13-0.40), and was highly correlated (P=.001) with pathologic response as opposed to biochemical response alone. After multivariate adjustment, metabolic response was the single largest independent preoperative predictor (P<.001) for pathologic response (odds ratio, 43.2; 95% CI, 16.9-153.2), RFS (hazard ratio, 0.37; 95% CI, 0.2-0.6), and OS (hazard ratio, 0.21; 95% CI, 0.1-0.4). CONCLUSIONS Among patients with post-NAT resected BR/LA PDAC, FDG-PET highly predicts pathologic response and survival, superior to biochemical responses alone. Given the poor ability of anatomic imaging or biochemical markers to assess NAT responses in these patients, FDG-PET is a preoperative metric of NAT efficacy, thereby allowing potential therapeutic alterations and surgical treatment decisions. We suggest that FDG-PET should be an adjunct and recommended modality during the NAT phase of care for these patients.
Collapse
Affiliation(s)
| | - Ajit H. Goenka
- Division of Nuclear Medicine Radiology, Department of Radiology
| | - Roberto Alva-Ruiz
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| | | | | | - Rondell P. Graham
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology
| | | | | | | | - Susanne G. Warner
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| | | | - Travis E. Grotz
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| | - Nguyen H. Tran
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Rory L. Smoot
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| | - Wen Wee Ma
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Sean P. Cleary
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| | - Robert R. McWilliams
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - David M. Nagorney
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| | | | | | - Mark J. Truty
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| |
Collapse
|
29
|
Seo YD, Katz MHG. Preoperative therapy for pancreatic adenocarcinoma-precision beyond anatomy. Cancer 2022; 128:3041-3056. [PMID: 35679197 DOI: 10.1002/cncr.34273] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 12/16/2022]
Abstract
Despite recent advances in the systemic treatment of gastrointestinal tumors, pancreatic adenocarcinoma (PDAC) remains a challenging disease, with 5-year survival just over 10%. Pancreatectomy in patients meeting defined anatomic criteria can result in cure; however, perioperative morbidity and mortality, as well as high rates of both local and distant recurrence even after "potentially curative" resection, have limited survival. Although perioperative chemotherapy has been shown to improve patients' longevity and chance for cure, debate continues about whether the preoperative or adjuvant approach is most effective in treatment of localized PDAC. Large, randomized multicenter trials in patients with resectable and borderline resectable PDAC have evaluated an evolving therapeutic landscape with mixed results. Importantly, these landmark studies share the fundamentally flawed assumption that tumor anatomical characteristics are an indicator of behavior and natural history. Concurrent biologic and translational research has revealed that rather than a single disease, PDAC represents a phenotypically variable group of malignancies arising in physiologically diverse patients. Ongoing novel trials have begun to capture this heterogeneity both in patient selection as well as the measurement of response by using genomic, transcriptional and radiomic markers. By moving away from classic anatomic descriptors to a more nuanced landscape of biomarkers predictive of tumor behavior and response, we can further refine the questions asked in preoperative trials and translate the answers to clinically meaningful precision therapy in localized PDAC.
Collapse
Affiliation(s)
- Y David Seo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
30
|
Oba A, Del Chiaro M, Satoi S, Kim SW, Takahashi H, Yu J, Hioki M, Tanaka M, Kato Y, Ariake K, Wu YHA, Inoue Y, Takahashi Y, Hackert T, Wolfgang CL, Besselink MG, Schulick RD, Nagakawa Y, Isaji S, Tsuchida A, Endo I. New criteria of resectability for pancreatic cancer: A position paper by the Japanese Society of Hepato-Biliary-Pancreatic Surgery (JSHBPS). JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2022; 29:725-731. [PMID: 34581016 DOI: 10.1002/jhbp.1049] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/28/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022]
Abstract
The symposium "New criteria of resectability for pancreatic cancer" was held during the 33nd meeting of the Japanese Society of Hepato-Biliary-Pancreatic Surgery (JSHBPS) in 2021 to discuss the potential modifications that could be made in the current resectability classification. The meeting focused on setting the foundation for developing a new prognosis-based resectability classification that is based on the tumor biology and the response to neoadjuvant treatment (NAT). The symposium included selected experts from Western and Eastern high-volume centers who have discussed their concept of resectability status through published literature. During the symposium, presenters reported new resectability classifications from their respective institutions based on tumor biology, conditional status, pathology, and genetics, in addition to anatomical tumor involvement. Interestingly, experts from all the centers reached the agreement that anatomy alone is insufficient to define resectability in the current era of effective NAT. On behalf of the JSHBPS, we would like to summarize the content of the conference in this position paper. We also invite global experts as internal reviewers of this paper for intercontinental cooperation in creating an up-to-date, prognosis-based resectability classification that reflects the trends of contemporary clinical practice.
Collapse
Affiliation(s)
- Atsushi Oba
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Marco Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sohei Satoi
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
- Department Surgery, Kansai Medical University, Osaka, Japan
| | - Sun-Whe Kim
- Department Surgery, Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang, Korea
| | - Hidenori Takahashi
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Jun Yu
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Masayoshi Hioki
- Department of Surgery, Fukuyama City Hospital, Hiroshima, Japan
| | - Masayuki Tanaka
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Germany
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiyasu Kato
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kyohei Ariake
- Department of Surgery, Tohoku University, Sendai, Japan
| | - Y H Andrew Wu
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Yosuke Inoue
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yu Takahashi
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Germany
| | | | - Marc G Besselink
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Richard D Schulick
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Yuichi Nagakawa
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, Tokyo, Japan
| | - Shuji Isaji
- Director of Mie University Graduate School of Medicine, Tsu, Japan
| | - Akihiko Tsuchida
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, Tokyo, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School Medicine, Yokohama, Japan
| |
Collapse
|
31
|
Tabata K, Nishie A, Shimomura Y, Isoda T, Kitamura Y, Nakata K, Yamada Y, Oda Y, Ishigami K, Baba S. Prediction of pathological response to preoperative chemotherapy for pancreatic ductal adenocarcinoma using 2-[ 18F]-fluoro-2-deoxy-d-glucose positron-emission tomography. Clin Radiol 2022; 77:436-442. [PMID: 35410786 DOI: 10.1016/j.crad.2022.03.001] [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: 11/05/2021] [Accepted: 03/02/2022] [Indexed: 12/24/2022]
Abstract
AIM To determine whether the pathological response to preoperative chemotherapy for pancreatic ductal adenocarcinoma (PDAC) can be predicted using 2-[18F]-fluoro-2-deoxy-d-glucose positron-emission tomography (F-18 FDG-PET). MATERIALS AND METHODS Twenty-eight patients with PDAC who underwent only neoadjuvant chemotherapy (NAC) before surgery were enrolled in the study. All patients had F-18 FDG-PET examinations before NAC. The resected specimen was pathologically evaluated according to the Classification of Pancreatic Carcinoma (7th edn). Patients were categorised into a non-response group and a response group based on the pathological findings. The non-response group (Grades 1a and 1b) showed ≤50% necrosis in the specimen, while the specimens of the response group (Grades 2-3) showed >50% necrosis. The maximum standardised uptake values (SUVmax) of the tumours on F-18 FDG-PET were measured. The mean values of SUVmax were compared between the two groups. The diagnostic performance of SUVmax in distinguishing the two groups was also evaluated using receiver operating characteristic analysis. RESULTS The mean SUVmax of the response group was higher than that of the non-response group (9.00 ± 1.78 versus 4.26 ± 2.35; p<0.001). The optimal cut-off value of SUVmax was 9.28 for distinguishing the two groups. The sensitivity, specificity, and accuracy for the prediction in the response group were 80%, 95.7%, and 92.9%, respectively. CONCLUSIONS SUVmax on F-18 FDG-PET may be useful as a biomarker to predict the pathological response to NAC in patients with PDAC.
Collapse
Affiliation(s)
- K Tabata
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku Fukuoka, 812-8582, Japan
| | - A Nishie
- Department of Radiology Informatics and Network, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku Fukuoka, 812-8582, Japan; Department of Radiology, Graduate School of Medical Science, University of the Ryukyus, 207, Uehara, Nishihara-cho, Okinawa, 903-0215, Japan.
| | - Y Shimomura
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku Fukuoka, 812-8582, Japan
| | - T Isoda
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku Fukuoka, 812-8582, Japan
| | - Y Kitamura
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku Fukuoka, 812-8582, Japan
| | - K Nakata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku Fukuoka, 812-8582, Japan
| | - Y Yamada
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku Fukuoka, 812-8582, Japan
| | - Y Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku Fukuoka, 812-8582, Japan
| | - K Ishigami
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku Fukuoka, 812-8582, Japan
| | - S Baba
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku Fukuoka, 812-8582, Japan
| |
Collapse
|
32
|
Qu C, Zeng PE, Wang HY, Yuan CH, Yuan HS, Xiu DR. Application of Magnetic Resonance Imaging in Neoadjuvant Treatment of Pancreatic Ductal Adenocarcinoma. J Magn Reson Imaging 2022; 55:1625-1632. [PMID: 35132729 DOI: 10.1002/jmri.28096] [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: 12/09/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignant tumors of the human digestive system. Due to its insidious onset, many patients have already lost the opportunity for radical resection upon tumor diagnosis. In recent years, neoadjuvant treatment for patients with borderline resectable PDAC has been recommended by multiple guidelines to increase the resection rate of radical surgery and improve the postoperative survival. However, further developments are required to accurately assess the tumor response to neoadjuvant therapy and to select the population suitable for such treatment. Reductions in drug toxicity and the number of neoadjuvant cycles are also critical. At present, the clinical evaluation of neoadjuvant treatment is mainly based on several serological and imaging indicators; however, the unique characteristics of PDAC and the insufficient sensitivity and specificity of the markers render this system ineffective. The imaging evaluation system, magnetic resonance imaging (MRI), has its own unique imaging advantages compared with computed tomography (CT) and other imaging examinations. One key advantage is the ability to reflect the changes more rapidly in tumor tissue components, such as the degree of fibrosis, microvessel density, and tissue hypoxia. It can also perform multiparameter quantitative analysis of tumor tissue and changes, attributing to its increasingly important role in imaging evaluation, and potentially the evaluation of neoadjuvant treatment of pancreatic cancer, as several current articles have studied. At the same time, owing to the complexity of MRI and some of its limitations, its wider application is limited. Compared with CT imaging, few relevant studies have been conducted. In this review article, we will investigate and summarize the advantages, limitations, and future development of MRI in the evaluation of neoadjuvant treatment of PDAC. EVIDENCE LEVEL: 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Chao Qu
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Piao-E Zeng
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Hang-Yan Wang
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Chun-Hui Yuan
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Hui-Shu Yuan
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Dian-Rong Xiu
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| |
Collapse
|
33
|
Labori KJ. Short-Course or Total Neoadjuvant Chemotherapy in Resectable and Borderline Resectable Pancreatic Cancer - Current Status and Future Perspectives. Front Surg 2022; 9:839339. [PMID: 35548190 PMCID: PMC9082635 DOI: 10.3389/fsurg.2022.839339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/28/2022] [Indexed: 12/27/2022] Open
Abstract
Neoadjuvant therapy improves overall survival compared with a surgery-first approach in patients with borderline resectable pancreatic cancer (BRPC). Evidence of higher quality is required to determine whether neoadjuvant therapy has potential benefits and improves survival for patients with resectable pancreatic cancer (RPC). Most randomized controlled trials (RCTs) have explored short-course neoadjuvant chemotherapy (SNT), but total neoadjuvant chemotherapy (TNT) is now the experimental arm of ongoing RCTs. This article reviews the current status of SNT and TNT in RPC and BRPC, and provides perspectives of future challenges and research directions in this field.
Collapse
Affiliation(s)
- Knut Jørgen Labori
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
34
|
Blair AB, Gemenetzis G, He J. The Role of Diffusion-Weighted Magnetic Resonance Imaging in Staging After Neoadjuvant Chemotherapy in Locally Advanced Pancreatic Adenocarcinoma: Reply. Ann Surg Oncol 2022; 29:10.1245/s10434-022-11737-4. [PMID: 35437667 DOI: 10.1245/s10434-022-11737-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 03/28/2022] [Indexed: 11/18/2022]
Affiliation(s)
- Alex B Blair
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Georgios Gemenetzis
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Surgery, Royal Infirmary Edinburgh, Edinburgh, Scotland, UK
| | - Jin He
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
35
|
Alva-Ruiz R, Yohanathan L, Yonkus JA, Abdelrahman AM, Gregory LA, Halfdanarson TR, Mahipal A, McWilliams RR, Ma WW, Hallemeier CL, Graham RP, Grotz TE, Smoot RL, Cleary SP, Nagorney DM, Kendrick ML, Truty MJ. Neoadjuvant Chemotherapy Switch in Borderline Resectable/Locally Advanced Pancreatic Cancer. Ann Surg Oncol 2022; 29:1579-1591. [PMID: 34724125 PMCID: PMC8810469 DOI: 10.1245/s10434-021-10991-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/06/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Neoadjuvant chemotherapy (NAC) is an integral part of preoperative treatment for patients with borderline resectable/locally advanced (BR/LA) pancreatic ductal adenocarcinoma (PDAC). The identification of a chemotherapeutic regimen that is both effective and tolerable is critical for NAC to be of oncologic benefit. After initial first-line (FL) NAC, some patients have lack of response or therapeutic toxicities precluding further treatment with the same regimen; optimal decision making regarding this patient population is unclear. Chemotherapy switch (CS) may allow for a larger proportion of patients to undergo curative-intent resection after NAC. METHODS We reviewed our surgical database for patients undergoing combinatorial NAC for BR/LA PDAC. Variant histologic exocrine carcinomas, intraductal papillary mucinous neoplasm-associated PDAC, and patients without research consent were excluded. RESULTS Overall, 468 patients with BR/LA PDAC receiving FL chemotherapy were reviewed, of whom 70% (329/468) continued with FL chemotherapy followed by surgical resection. The remaining 30% (139/468) underwent CS, with 72% (100/139) of CS patients going on to curative-intent surgical resection. Recurrence-free survival (RFS) and overall survival (OS) were not significantly different between the resected FL and CS cohorts (30.0 vs. 19.1 months, p = 0.13, and 41.4 vs. 36.4 months, p = 0.94, respectively) and OS was significantly worse in those undergoing CS without subsequent resection (19 months, p < 0.0001). On multivariable analysis, carbohydrate antigen (CA) 19-9 and pathologic treatment responses were predictors of RFS and OS. CONCLUSION CS in patients undergoing NAC for BR/LA pancreatic cancer does not incur oncologic detriment. The incorporation of CS into NAC treatment sequencing may allow a greater proportion of patients to proceed to curative-intent surgery.
Collapse
Affiliation(s)
- Roberto Alva-Ruiz
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Lavanya Yohanathan
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Jennifer A Yonkus
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Amro M Abdelrahman
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Lindsey A Gregory
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Amit Mahipal
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | | | - Wen Wee Ma
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | | | - Rondell P Graham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Travis E Grotz
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Rory L Smoot
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Sean P Cleary
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - David M Nagorney
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Michael L Kendrick
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Mark J Truty
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
36
|
Boggi U, Truty M, Zyromski NJ. 2021 SSAT Debate: Selective Approach to Resection of the Superior Mesenteric Artery in Pancreatic Cancer vs Superior Mesenteric Artery Encasement Is Not an Absolute Contraindication for Surgery in Pancreatic Cancer. J Gastrointest Surg 2022; 26:523-531. [PMID: 35059988 DOI: 10.1007/s11605-021-05237-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/03/2021] [Indexed: 02/08/2023]
Abstract
OVERVIEW This manuscript summarizes an excellent debate from the 2021 SSAT/Pancreas Club symposium on arterial resection in pancreas cancer. Two world-recognized experts, Professor Ugo Boggi from Pisa, IT, and Dr. Mark Truty from the Mayo Clinic in Rochester, MN, offered their views on the role of arterial resection in locally advanced pancreas ductal adenocarcinoma. Both speakers have extensive experience pushing the technical envelope with extended vascular resection in pancreatectomy. However, both highlight important concepts of resectability extending well beyond technique: namely, patient global physiology, tumor biology, and response to chemotherapy. The debate was spirited, and this subsequent review is an excellent look at the status quo. N. J. Zyromski, MD, Indianpolis, IN, November, 2021.
Collapse
|
37
|
van Dam MA, Vuijk FA, Stibbe JA, Houvast RD, Luelmo SAC, Crobach S, Shahbazi Feshtali S, de Geus-Oei LF, Bonsing BA, Sier CFM, Kuppen PJK, Swijnenburg RJ, Windhorst AD, Burggraaf J, Vahrmeijer AL, Mieog JSD. Overview and Future Perspectives on Tumor-Targeted Positron Emission Tomography and Fluorescence Imaging of Pancreatic Cancer in the Era of Neoadjuvant Therapy. Cancers (Basel) 2021; 13:6088. [PMID: 34885196 PMCID: PMC8656821 DOI: 10.3390/cancers13236088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/25/2021] [Accepted: 11/28/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Despite recent advances in the multimodal treatment of pancreatic ductal adenocarcinoma (PDAC), overall survival remains poor with a 5-year cumulative survival of approximately 10%. Neoadjuvant (chemo- and/or radio-) therapy is increasingly incorporated in treatment strategies for patients with (borderline) resectable and locally advanced disease. Neoadjuvant therapy aims to improve radical resection rates by reducing tumor mass and (partial) encasement of important vascular structures, as well as eradicating occult micrometastases. Results from recent multicenter clinical trials evaluating this approach demonstrate prolonged survival and increased complete surgical resection rates (R0). Currently, tumor response to neoadjuvant therapy is monitored using computed tomography (CT) following the RECIST 1.1 criteria. Accurate assessment of neoadjuvant treatment response and tumor resectability is considered a major challenge, as current conventional imaging modalities provide limited accuracy and specificity for discrimination between necrosis, fibrosis, and remaining vital tumor tissue. As a consequence, resections with tumor-positive margins and subsequent early locoregional tumor recurrences are observed in a substantial number of patients following surgical resection with curative intent. Of these patients, up to 80% are diagnosed with recurrent disease after a median disease-free interval of merely 8 months. These numbers underline the urgent need to improve imaging modalities for more accurate assessment of therapy response and subsequent re-staging of disease, thereby aiming to optimize individual patient's treatment strategy. In cases of curative intent resection, additional intra-operative real-time guidance could aid surgeons during complex procedures and potentially reduce the rate of incomplete resections and early (locoregional) tumor recurrences. In recent years intraoperative imaging in cancer has made a shift towards tumor-specific molecular targeting. Several important molecular targets have been identified that show overexpression in PDAC, for example: CA19.9, CEA, EGFR, VEGFR/VEGF-A, uPA/uPAR, and various integrins. Tumor-targeted PET/CT combined with intraoperative fluorescence imaging, could provide valuable information for tumor detection and staging, therapy response evaluation with re-staging of disease and intraoperative guidance during surgical resection of PDAC. METHODS A literature search in the PubMed database and (inter)national trial registers was conducted, focusing on studies published over the last 15 years. Data and information of eligible articles regarding PET/CT as well as fluorescence imaging in PDAC were reviewed. Areas covered: This review covers the current strategies, obstacles, challenges, and developments in targeted tumor imaging, focusing on the feasibility and value of PET/CT and fluorescence imaging for integration in the work-up and treatment of PDAC. An overview is given of identified targets and their characteristics, as well as the available literature of conducted and ongoing clinical and preclinical trials evaluating PDAC-targeted nuclear and fluorescent tracers.
Collapse
Affiliation(s)
- Martijn A. van Dam
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Floris A. Vuijk
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Judith A. Stibbe
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Ruben D. Houvast
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Saskia A. C. Luelmo
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Stijn Crobach
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | | | - Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, University Medical Center Leiden, 2333 ZA Leiden, The Netherlands;
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, The Netherlands
| | - Bert A. Bonsing
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Cornelis F. M. Sier
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
- Percuros B.V., 2333 CL Leiden, The Netherlands
| | - Peter J. K. Kuppen
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | | | - Albert D. Windhorst
- Department of Radiology, Section of Nuclear Medicine, Amsterdam UMC, Location VUmc, 1081 HV Amsterdam, The Netherlands;
| | - Jacobus Burggraaf
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
- Centre for Human Drug Research, 2333 CL Leiden, The Netherlands
| | - Alexander L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - J. Sven D. Mieog
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| |
Collapse
|
38
|
Do-It-Yourself Business Intelligence for the Radiologist-Lessons Learned From 10-Year Trends in an Abdominal Imaging Division at a Tertiary Medical Center. J Am Coll Radiol 2021; 19:329-335. [PMID: 34852270 DOI: 10.1016/j.jacr.2021.10.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/08/2021] [Indexed: 12/26/2022]
Abstract
"Big data" has changed the way businesses operate in finance, insurance, communications, manufacturing, and logistics, with companies reporting successful outcomes from the implementation of data-driven analytics. Health care has slowly started leveraging big data analysis to identify more cost-efficient and effective care for patients. Business intelligence software aims to help with this transition, converting large data sets into useful, actionable information. Radiology practices have used business intelligence toolkits in the past to increase reading room productivity, scanner efficiency, and patient throughput. Although these tactics have driven down costs and increased volumes, an essential aspect of radiology practice growth-consumer satisfaction-remains largely unexplored. The first commandment of marketing is "know thy customer," which is the key to increasing radiology's value to referring clinicians who use our services. Our article explores the business case behind a basic business intelligence model, giving any radiologist the instruments to access and draw business conclusions from their radiology information system data.
Collapse
|
39
|
Vascular resections in minimally invasive surgery for pancreatic cancer. LAPAROSCOPIC, ENDOSCOPIC AND ROBOTIC SURGERY 2021. [DOI: 10.1016/j.lers.2021.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
40
|
Editor's Notebook: September 2021. AJR Am J Roentgenol 2021; 217:527-528. [PMID: 34415203 DOI: 10.2214/ajr.21.26336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
41
|
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
|
42
|
Benz MR, Armstrong WR, Ceci F, Polverari G, Donahue TR, Wainberg ZA, Quon A, Auerbach M, Girgis MD, Herrmann K, Czernin J, Calais J. 18F-FDG PET/CT imaging biomarkers for early and late evaluation of response to first-line chemotherapy in patients with pancreatic ductal adenocarcinoma. J Nucl Med 2021; 63:199-204. [PMID: 34272317 DOI: 10.2967/jnumed.121.261952] [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: 01/20/2021] [Revised: 05/05/2021] [Indexed: 11/16/2022] Open
Abstract
Purpose: The purpose of this study was to evaluate 18F-FDG-PET/CT as an early and late interim imaging biomarker in patients with pancreatic ductal adenocarcinoma (PDAC) who undergo first-line systemic therapy. Methods: This was a prospective, single-center, single-arm, open-label study (IRB12-000770). Patient receiving first line chemotherapy were planned to undergo a baseline 18F-FDG-PET/CT (PET1), early interim 18F-FDG PET/CT (PET2) and late interim 18F-FDG-PET/CT (PET3). ROC selected and established (mPERCIST / RECIST1.1) cut-offs for metabolic and radiographic tumor response assessment were applied. Patients were followed to collect data on further treatments and overall survival (OS). Results: The study population consisted of 28 patients who underwent PET1. Twenty-three of these (82%) underwent PET2 and 21 (75%) PET3, respectively. Twenty-three deaths occurred during a median follow up period of 14 months (maximum follow up, 58.3 months). The median OS was 36.2 months (95%CI, 28-NYR) in early metabolic responders (6/23 (26%), P = 0.016) and 25.4 months (95%CI, 19.6-NYR) in early radiographic responders (7/23 (30%), P = 0.16). The median overall survival was 27.4 months (95%CI, 21.4-NYR) in late metabolic responders (10/21 (48%), P = 0.058) and 58.2 months (95%CI, 21.4-NYR) in late radiographic responders (7/21 (33%), P = 0.008). Conclusion: 18F-FDG PET may serve as early interim imaging biomarker (~ at 4 weeks) for evaluation of response to first-line chemotherapy in patients with PDAC. Radiographic changes might be sufficient for response evaluation after the completion of first line chemotherapy.
Collapse
Affiliation(s)
- Matthias R Benz
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, United States
| | - Wesley R Armstrong
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, United States
| | - Francesco Ceci
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, Milan, Italy
| | | | | | - Zev A Wainberg
- Department of Medical Oncology, University of California, Los Angeles, CA
| | - Andrew Quon
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, United States
| | - Martin Auerbach
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, United States
| | - Mark D Girgis
- Department of Surgery, University of California, Los Angeles, CA
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Johannes Czernin
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, United States
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, United States
| |
Collapse
|
43
|
Evangelista L, Zucchetta P, Moletta L, Serafini S, Cassarino G, Pegoraro N, Bergamo F, Sperti C, Cecchin D. The role of FDG PET/CT or PET/MRI in assessing response to neoadjuvant therapy for patients with borderline or resectable pancreatic cancer: a systematic literature review. Ann Nucl Med 2021; 35:767-776. [PMID: 34047926 PMCID: PMC8197718 DOI: 10.1007/s12149-021-01629-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/11/2021] [Indexed: 02/05/2023]
Abstract
The aim of the present systematic review is to examine the role of fluorodeoxyglucose (FDG) positron emission tomography (PET) associated with computed tomography (CT) or magnetic resonance imaging (MRI) in assessing response to preoperative chemotherapy or chemoradiotherapy (CRT) for patients with borderline and resectable pancreatic ductal adenocarcinoma (PDAC). Three researchers ran a database query in PubMed, Web of Science and EMBASE. The total number of patients considered was 488. The most often used parameters of response to therapy were the reductions in the maximum standardized uptake value (SUVmax) or the peak standardized uptake lean mass (SULpeak). Patients whose SUVs were higher at the baseline (before CRT) were associated with a better response to therapy and a better overall survival. SUVs remaining high after neoadjuvant therapy correlated with a poor prognosis. Available data indicate that FDG PET/CT or PET/MRI can be useful for predicting and assessing response to CRT in patients with resectable or borderline PDAC.
Collapse
Affiliation(s)
- Laura Evangelista
- Nuclear Medicine Unit, Department of Medicine DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy.
| | - Pietro Zucchetta
- Nuclear Medicine Unit, Department of Medicine DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Lucia Moletta
- Department of Surgery, Oncology and Gastroenterology-DiSCOG, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Simone Serafini
- Department of Surgery, Oncology and Gastroenterology-DiSCOG, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Gianluca Cassarino
- Nuclear Medicine Unit, Department of Medicine DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Nicola Pegoraro
- Nuclear Medicine Unit, Department of Medicine DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Francesca Bergamo
- Medical Oncology Unit 1, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128, Padua, Italy
| | - Cosimo Sperti
- Department of Surgery, Oncology and Gastroenterology-DiSCOG, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Diego Cecchin
- Nuclear Medicine Unit, Department of Medicine DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| |
Collapse
|
44
|
Zhang Y, Huang ZX, Song B. Role of imaging in evaluating the response after neoadjuvant treatment for pancreatic ductal adenocarcinoma. World J Gastroenterol 2021; 27:3037-3049. [PMID: 34168406 PMCID: PMC8192284 DOI: 10.3748/wjg.v27.i22.3037] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/08/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy. Despite the development of multimodality treatments, including surgical resection, radiotherapy, and chemotherapy, the long-term prognosis of patients with PDAC remains poor. Recently, the introduction of neoadjuvant treatment (NAT) has made more patients amenable to surgery, increasing the possibility of R0 resection, treatment of occult micro-metastasis, and prolongation of overall survival. Imaging plays a vital role in tumor response evaluation after NAT. However, conventional imaging modalities such as multidetector computed tomography have limited roles in the assessment of tumor resectability after NAT for PDAC because of the similar appearance of tissue fibrosis and tumor infiltration. Perfusion computed tomography, using blood perfusion as a biomarker, provides added value in predicting the histopathologic response of PDAC to NAT by reflecting the changes in tumor matrix and fibrosis content. Other imaging technologies, including diffusion-weighted imaging of magnetic resonance imaging and positron emission tomography, can reveal the tumor response by monitoring the structural changes in tumor cells and functional metabolic changes in tumors after NAT. In addition, with the renewed interest in data acquisition and analysis, texture analysis and radiomics have shown potential for the early evaluation of the response to NAT, thus improving patient stratification to achieve accurate and intensive treatment. In this review, we briefly introduce the application and value of NAT in resectable and unresectable PDAC. We also summarize the role of imaging in evaluating the response to NAT for PDAC, as well as the advantages, limitations, and future development directions of current imaging techniques.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Zi-Xing Huang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|