101
|
Luan X, Wang X, Bian G, Li X, Gao Z, Liu Z, Zhang Z, Han T, Zhao J, Zhao H, Luan X, Zhu W, Dong L, Guo F. Exosome applications for the diagnosis and treatment of pancreatic ductal adenocarcinoma: An update (Review). Oncol Rep 2025; 53:13. [PMID: 39575479 PMCID: PMC11605277 DOI: 10.3892/or.2024.8846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a malignant neoplasm that typically manifests with subtle clinical manifestations in its early stages and frequently eludes diagnosis until the advanced phases of the disease. The limited therapeutic options available for PDAC significantly contribute to its high mortality rate, highlighting the urgent need for novel biomarkers capable of effectively identifying early clinical manifestations and facilitating precise diagnosis. The pivotal role of cellular exosomes in both the pathogenesis and therapeutic interventions for PDAC has been underscored. Furthermore, researchers have acknowledged the potential of exosomes as targeted drug carriers against regulatory cells in treating PDAC. The present article aims to provide a comprehensive review encompassing recent advancements in utilizing exosomes for elucidating mechanisms underlying disease development, patterns of metastasis, diagnostic techniques and treatment strategies associated with PDAC.
Collapse
Affiliation(s)
- Xinchi Luan
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Xuezhe Wang
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Gang Bian
- Department of Gastroenterology, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, Shandong 266041, P.R. China
| | - Xiaoxuan Li
- Department of Oncology, Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266031, P.R. China
| | - Ziru Gao
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Zijiao Liu
- School of Clinical and Basic Medicine and Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Zhishang Zhang
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Tianyue Han
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Jinpeng Zhao
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Hongjiao Zhao
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Xinyue Luan
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Wuhui Zhu
- Department of Hepatobiliary surgery, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, Shandong 266041, P.R. China
| | - Lili Dong
- Department of Gastroenterology, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, Shandong 266041, P.R. China
| | - Feifei Guo
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
102
|
Matsumoto M, Uwagawa T, Shirai Y, Tsunematsu M, Furukawa K, Haruki K, Okui N, Okazaki K, Ishizaki S, Ikegami T. Impact of Preoperative Osteosarcopenia and Postoperative Administration of Pancrelipase on the Prognosis of Borderline Resectable and Unresectable Locally Advanced Pancreatic Cancer. Am Surg 2025; 91:65-75. [PMID: 39108198 DOI: 10.1177/00031348241272420] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
BACKGROUND This study aimed to identify postoperative recurrence and prognostic factors, including osteosarcopenia for borderline resectable (BR) and unresectable locally advanced (UR-LA) pancreatic cancer and to examine the impact of postoperative pancreatic enzyme replacement therapy (PERT). METHODS We retrospectively examined 32 resected patients with BR and UR-LA pancreatic cancer. We investigated independent factors in the disease-free survival and overall survival. The relation of osteosarcopenia with the clinicopathological factors was investigated. Additionally, the association of the administration of a standard dose of pancrelipase, the amount of lipase required for patients with pancreatic exocrine insufficiency, for ≥6 months postoperatively with improvement of sarcopenia, osteopenia, and osteosarcopenia and completion rate of adjuvant chemotherapy was investigated. RESULTS Multivariate analyses identified osteosarcopenia (P = 0.049) and lymph node metastasis (P = 0.01) as independent recurrence predictors, and osteosarcopenia (P = 0.002), maximum tumor diameter ≥40 mm (P = 0.006), and no adjuvant therapy (P = 0.01) as independent prognostic predictors. In the osteosarcopenia group, serum CA19-9 levels were higher (P = 0.03). The administration of a standard dose of pancrelipase for ≥6 months postoperatively was none in the osteosarcopenia group (0% vs 42.9%, P = 0.01), while significantly improved postoperative sarcopenia (33% vs 0%, P = 0.004), increased number of cycles of adjuvant chemotherapy (n = 6 vs n = 3, P = 0.03), and the completion rate of adjuvant chemotherapy in excluding cases interrupted because of recurrence (86% vs 25%, P = 0.007). CONCLUSIONS Osteosarcopenia was an independent recurrent and prognostic factor in patients after pancreatectomy for locally advanced pancreatic cancer. Appropriate postoperative PERT may contribute to a better prognosis by improving sarcopenia and increasing the completion rate of adjuvant chemotherapy.
Collapse
Affiliation(s)
- Michinori Matsumoto
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Tadashi Uwagawa
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Yoshihiro Shirai
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Masashi Tsunematsu
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Kenei Furukawa
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Koichiro Haruki
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Norimitsu Okui
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Kohei Okazaki
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Shunta Ishizaki
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Toru Ikegami
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| |
Collapse
|
103
|
D'Ambra V, Ingaldi C, Ricci C, Alberici L, Capretti G, Jovine E, Zingaretti CC, Salvia R, Casadei R, Long-Survival SICO Study Group. Pancreatic cancer and long survivors: a survey of Italian society of oncological surgery (SICO). Updates Surg 2025; 77:57-64. [PMID: 39589628 DOI: 10.1007/s13304-024-02039-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/17/2024] [Indexed: 11/27/2024]
Abstract
Long-term survivors after pancreatic resection for PDAC are rare, constituting a specific subset of patients that remains poorly understood. The aim of this survey is to describe the current landscape related to survival in the Italian context and identify factors associated with long-term survival. An online survey, conducted by the Italian Society of Oncological Surgery (SICO) and endorsed by Italian Association of the Study of the Pancreas (AISP) and Italian Association of Hepatobiliary Pancreatic Surgery (AICEP), was distributed to surgeons in July 2023. The survey included 27 multiple-choice questions covering demographics, professional details, clinical practices, and long-term survival data. Responses were analyzed using descriptive statistics and multinomial logistic regression to identify factors related to long-term survival. The majority of surgeons (46.9%) considered LTS as "alive at 5 years, regardless of disease-free status". The percentage of patients alive at 5 years post-2013 was higher compared to pre-2013. Almost all centers (93.2%) held multidisciplinary discussions. Very high-volume centers (> 100 resections/year) in comparison to very low-volume (< 10 resections/year) showed better long-term survival rates. No difference in survival were observed between centers with low, medium, high, and very high volumes. In addition, centers with multidisciplinary approach showed better survival rates. Centers with more neoadjuvant chemotherapy rates, low-grade and low-stage tumors were also associated with improved survival outcomes. This survey has allowed to understand the Italian scenario regarding survival in patients undergoing surgery for PDAC.
Collapse
Affiliation(s)
- Vincenzo D'Ambra
- Division of Pancreatic Surgery, Department of Internal Medicine and Surgery (DIMEC), IRCCS, Azienda Ospedaliero Universitaria Di Bologna, Alma Mater Studiorum, University of Bologna, S. Orsola-Malpighi Hospital, Via Massarenti n.9, 40138, Bologna, Italy.
| | - Carlo Ingaldi
- Division of Pancreatic Surgery, Department of Internal Medicine and Surgery (DIMEC), IRCCS, Azienda Ospedaliero Universitaria Di Bologna, Alma Mater Studiorum, University of Bologna, S. Orsola-Malpighi Hospital, Via Massarenti n.9, 40138, Bologna, Italy
| | - Claudio Ricci
- Division of Pancreatic Surgery, Department of Internal Medicine and Surgery (DIMEC), IRCCS, Azienda Ospedaliero Universitaria Di Bologna, Alma Mater Studiorum, University of Bologna, S. Orsola-Malpighi Hospital, Via Massarenti n.9, 40138, Bologna, Italy
| | - Laura Alberici
- Division of Pancreatic Surgery, Department of Internal Medicine and Surgery (DIMEC), IRCCS, Azienda Ospedaliero Universitaria Di Bologna, Alma Mater Studiorum, University of Bologna, S. Orsola-Malpighi Hospital, Via Massarenti n.9, 40138, Bologna, Italy
| | - Giovanni Capretti
- Pancreatic Surgery Unit, Humanitas Clinical and Research Center-IRCCS, Via Manzoni 56, Rozzano, Milan, Italy
| | - Elio Jovine
- Department of General Surgery, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Maggiore Hospital, 40133, Bologna, Italy
| | - Caterina C Zingaretti
- Department of General and Oncological Surgery, Mauriziano Hospital "Umberto I", Largo Turati, 62, 10128, Turin, Italy
| | - Roberto Salvia
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Riccardo Casadei
- Division of Pancreatic Surgery, Department of Internal Medicine and Surgery (DIMEC), IRCCS, Azienda Ospedaliero Universitaria Di Bologna, Alma Mater Studiorum, University of Bologna, S. Orsola-Malpighi Hospital, Via Massarenti n.9, 40138, Bologna, Italy
| | | |
Collapse
Collaborators
Patrizia Marsanic, Ludovico Carbone, Domenico Tamburrino, Massimo Falconi, Francesco Izzo, Bruno Nardo, Nicola Cillara, Mario Pacilli, Serena Langella, Marco Angrisani, Mario Giuffrida, Gianluca Baiocchi, Ugo Boggi, Nicola Cinardi, Daniele Delogu, Riccardo Memeo, Marco Brizzolari, Lapo Bencini, Paolo Ubiali, Andrea Morini, Rosario Mina, Gabriella Lionetto, Alessandro Soave, Christian Cotsoglou, Marco Massani, Lorenzo Manzini, Carlo Gazia, Renato Patrone, Niccolò Napoli, Luca Moraldi, Giacomo Deiro, Silvana B Puglisi,
Collapse
|
104
|
Andel PCM, van Goor IWJM, Augustinus S, Berrevoet F, Besselink MG, Bhojwani R, Boggi U, Bouwense SAW, Cirkel GA, van Dam JL, Djanani A, Dorcaratto D, Dreyer S, den Dulk M, Frigerio I, Ghorbani P, Goetz MR, Groot Koerkamp B, Gryspeerdt F, Hidalgo Salinas C, Intven M, Izbicki JR, Jorba Martin R, Kauffmann EF, Klug R, Liem MSL, Luyer MDP, Maglione M, Martin-Perez E, Meerdink M, de Meijer VE, Nieuwenhuijs VB, Nikov A, Nunes V, Pando Rau E, Radenkovic D, Roeyen G, Sanchez-Bueno F, Serrablo A, Sparrelid E, Tepetes K, Thakkar RG, Tzimas GN, Verdonk RC, ten Winkel M, Zerbi A, Groot VP, Molenaar IQ, Daamen LA, van Santvoort HC. Routine Imaging or Symptomatic Follow-Up After Resection of Pancreatic Adenocarcinoma. JAMA Surg 2025; 160:74-84. [PMID: 39504033 PMCID: PMC11541741 DOI: 10.1001/jamasurg.2024.5024] [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: 06/26/2024] [Accepted: 08/29/2024] [Indexed: 11/09/2024]
Abstract
IMPORTANCE International guidelines lack consistency in their recommendations regarding routine imaging in the follow-up after pancreatic resection for pancreatic ductal adenocarcinoma (PDAC). Consequently, follow-up strategies differ between centers worldwide. OBJECTIVE To compare clinical outcomes, including recurrence-focused treatment and survival, in patients with PDAC recurrence who received symptomatic follow-up or routine imaging after pancreatic resection in international centers affiliated with the European-African Hepato-Pancreato-Biliary Association (E-AHPBA). DESIGN, SETTING, AND PARTICIPANTS This was a prospective, international, cross-sectional study. Patients from a total of 33 E-AHPBA centers from 13 countries were included between 2020 and 2021. According to the predefined study protocol, patients who underwent PDAC resection and were diagnosed with disease recurrence were prospectively included. Patients were stratified according to postoperative follow-up strategy: symptomatic follow-up (ie, without routine imaging) or routine imaging. EXPOSURES Symptomatic follow-up or routine imaging in patients who underwent PDAC resection. MAIN OUTCOMES AND MEASURES Overall survival (OS) was estimated with Kaplan-Meier curves and compared using the log-rank test. To adjust for potential confounders, multivariable logistic regression was used to evaluate the association between follow-up strategy and recurrence-focused treatment. Multivariable Cox proportional hazard analysis was used to study the independent association between follow-up strategy and OS. RESULTS Overall, 333 patients (mean [SD] age, 65 [11] years; 184 male [55%]) with PDAC recurrence were included. Median (IQR) follow-up at time of analysis 2 years after inclusion of the last patient was 40 (30-58) months. Of the total cohort, 98 patients (29%) received symptomatic follow-up, and 235 patients (71%) received routine imaging. OS was 23 months (95% CI, 19-29 months) vs 28 months (95% CI, 24-30 months) in the groups who received symptomatic follow-up vs routine imaging, respectively (P = .01). Routine imaging was associated with receiving recurrence-focused treatment (adjusted odds ratio, 2.57; 95% CI, 1.22-5.41; P = .01) and prolonged OS (adjusted hazard ratio, 0.75; 95% CI, 0.56-.99; P = .04). CONCLUSION AND RELEVANCE In this international, prospective, cross-sectional study, routine follow-up imaging after pancreatic resection for PDAC was independently associated with receiving recurrence-focused treatment and prolonged OS.
Collapse
Affiliation(s)
- Paul C. M. Andel
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
- St Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands
| | - Iris W. J. M. van Goor
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Simone Augustinus
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Frederik Berrevoet
- Department of General and HPB Surgery and Liver Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Marc G. Besselink
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Rajesh Bhojwani
- Department of Surgery, Santokba Institute of Digestive Surgical Sciences, Santokba Durlabhji Memorial Hospital, Rajasthan, India
| | - Ugo Boggi
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Stefan A. W. Bouwense
- Department of Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Surgery, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Geert A. Cirkel
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jacob L. van Dam
- Department of Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Angela Djanani
- Department of Internal Medicine, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Dimitri Dorcaratto
- Department of Surgery, Hospital Clínico, University of Valencia, Biomedical Research Institute (INCLIVA), Valencia, Spain
| | - Stephan Dreyer
- Department of Academic Surgery, Glasgow Royal Infirmary, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Marcel den Dulk
- Department of Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Surgery, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Isabella Frigerio
- Pancreatic Surgical Unit, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Poya Ghorbani
- Department of Surgery, Karolinska Institutet, Solna, Sweden
| | - Mara R. Goetz
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg, Hamburg, Germany
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Filip Gryspeerdt
- Department of General and HPB Surgery and Liver Transplantation, Ghent University Hospital, Ghent, Belgium
| | | | - Martijn Intven
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jakob R. Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg, Hamburg, Germany
| | - Rosa Jorba Martin
- Department of Surgery, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
| | - Emanuele F. Kauffmann
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Reinhold Klug
- Department of General-, Visceral- and Vascular Surgery, Community Hospital Horn, Horn, Austria
| | - Mike S. L. Liem
- Department of Surgery, Medical Spectrum Twente, Enschede, the Netherlands
| | - Misha D. P. Luyer
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands
| | - Manuel Maglione
- Department of Visceral, Transplant, and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Elena Martin-Perez
- Department of General and Digestive Surgery, University Hospital La Princesa, Madrid, Spain
| | - Mark Meerdink
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Vincent E. de Meijer
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | | | - Andrej Nikov
- Department of Surgery, Military University Hospital Prague, Prague, Czech Republic
| | - Vitor Nunes
- Department of Surgery, Hospital Prof Doutor Fernando Fonseca EPE, Amadora, Portugal
| | - Elizabeth Pando Rau
- Department of Hepato-Pancreatobiliary and Transplant Surgery, Hospital Vall d’Hebron, Barcelona, Spain
- Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Dejan Radenkovic
- Clinic for Digestive Surgery, University Clinical Centra of Serbia, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Geert Roeyen
- Department of HPB, Endocrine and Transplantation Surgery, Antwerp University Hospital, Antwerp, Belgium
| | - Francisco Sanchez-Bueno
- Department of General and Digestive Surgery, Hospital Clínico Universitario “Virgen de la Arrixaca,” Murcia, Spain
| | - Alejandro Serrablo
- Department of General and Digestive Surgery, Miguel Servet University Hospital, Zaragoza, Spain
| | | | | | - Rohan G. Thakkar
- Department of Surgery, Newcastle Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - George N. Tzimas
- Department of HepatoPancreatoBiliary Surgery, Hygeia Hospital, Marousi, Greece
| | - Robert C. Verdonk
- Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Department of Gastroenterology, Utrecht, the Netherlands
| | | | - Alessandro Zerbi
- Department of Pancreatic Surgery, IRCCS Humanitas Hospital, Rozzano, and Humanitas University, Pieve Emanuele, Italy
| | - Vincent P. Groot
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| | - I. Quintus Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| | - Lois A. Daamen
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hjalmar C. van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| |
Collapse
|
105
|
Larsson P, Swartling O, Cheraghi D, Khawaja A, Soreide K, Sparrelid E, Ghorbani P. Assessment of Outcomes by Intention-to-Treat Comparison for Locally Advanced Pancreatic Cancer: A Population-Derived Cohort Study. Ann Surg Oncol 2025; 32:508-516. [PMID: 39365549 DOI: 10.1245/s10434-024-16291-9] [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: 07/09/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND The overall treatment response among patients with locally advanced pancreatic cancer (LAPC) is poorly understood as most studies report solely on resected patients. We aimed to investigate the outcomes in patients with LAPC as an intention-to-treat-analysis from the time of diagnosis from a complete source population. PATIENTS AND METHODS An observational cohort study in a population-defined region within a universal healthcare system. All consecutive patients discussed at multi-disciplinary tumour board (MDT), aged ≥ 18 years and diagnosed with LAPC were included. Exposure was set as recommended treatment by MDT (i.e. upfront surgery, neoadjuvant therapy, palliative treatment or best supportive care). Outcome measures were overall survival analysed by Kaplan-Meier survival estimates and multivariable analyses using logistic regression for odds ratios (OR) and Cox proportional hazard analysis for hazard ratios (HR). RESULTS In total, 8803 MDT events (6055 unique patients) with pancreatic disease were held during the study period. Some 1436 (24%) had pancreatic cancer, of which 162 (11%) had LAPC and 134 met the population-defined criteria. In overall survival analyses, the patients who were recommended neoadjuvant therapy (± surgery) demonstrated no significant difference to palliative chemotherapy (median 11.0 months vs. 11.8 months; p = 0.226). In multivariable analysis, adjusted OR for overall survival comparing the treatment groups was 0.27 (95% CI 0.02-3.29, p = 0.306) and Cox proportional HR 0.96 (95% CI 0.58-1.59, p = 0.865). CONCLUSIONS In patients with LAPC, survival was not statistically different between those recommended for attempt at neoadjuvant (± surgery) compared with those recommended palliative chemotherapy. The findings suggest that conversion/downstaging chemotherapy is successful in only a select few.
Collapse
Affiliation(s)
- Patrik Larsson
- Division of Surgery and Oncology, Department of Clinical Sciences, Interventions and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Oskar Swartling
- Division of Surgery and Oncology, Department of Clinical Sciences, Interventions and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Diana Cheraghi
- Division of Surgery and Oncology, Department of Clinical Sciences, Interventions and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ajnon Khawaja
- Division of Surgery and Oncology, Department of Clinical Sciences, Interventions and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Kjetil Soreide
- Division of Surgery and Oncology, Department of Clinical Sciences, Interventions and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ernesto Sparrelid
- Division of Surgery and Oncology, Department of Clinical Sciences, Interventions and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Poya Ghorbani
- Division of Surgery and Oncology, Department of Clinical Sciences, Interventions and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
106
|
Daibo S, Homma Y, Ohya H, Fukuoka H, Miyake K, Ozawa M, Kumamoto T, Matsuyama R, Saigusa Y, Endo I. Novel machine-learning model for predicting lymph node metastasis in resectable pancreatic ductal adenocarcinoma. Ann Gastroenterol Surg 2025; 9:161-168. [PMID: 39759999 PMCID: PMC11693540 DOI: 10.1002/ags3.12836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/14/2024] [Accepted: 06/03/2024] [Indexed: 01/07/2025] Open
Abstract
Aim Lymph node metastasis is an adverse prognostic factor in pancreatic ductal adenocarcinoma. However, it remains a challenge to predict lymph node metastasis using preoperative imaging alone. We used machine learning (combining preoperative imaging findings, tumor markers, and clinical information) to create a novel prediction model for lymph node metastasis in resectable pancreatic ductal adenocarcinoma. Methods The data of patients with resectable pancreatic ductal adenocarcinoma who underwent surgery between September 1991 and October 2022 were retrospectively examined. Machine-learning software (Statistical Package for the Social Sciences Modeler) was used to create a prediction model, and parameter tuning was performed to improve the model's accuracy. We also analyzed the contribution of each feature to prediction using individual conditional expectation and partial dependence plots. Results Of the 331 cases included in the study, 241 comprised the training cohort and 90 comprised the test cohort. After parameter tuning, the areas under the receiver operating characteristic curves for the training and test cohorts were 0.780 and 0.795, respectively. Individual conditional expectation and partial dependence plots showed that larger tumor size and carbohydrate antigen 19-9 and Duke pancreatic monoclonal antigen type 2 levels were associated with positive lymph node metastasis prediction in this model; neoadjuvant treatment was associated with negative lymph node metastasis prediction. Conclusion Machine learning may contribute to the creation of an effective predictive model of lymph node metastasis in pancreatic ductal adenocarcinoma. Prediction models using machine learning may contribute to the development of new treatment strategies in resectable pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Susumu Daibo
- Department of Gastroenterological SurgeryYokohama City UniversityYokohamaKanagawaJapan
| | - Yuki Homma
- Department of Gastroenterological SurgeryYokohama City UniversityYokohamaKanagawaJapan
| | - Hiroki Ohya
- Department of Gastroenterological SurgeryYokohama City UniversityYokohamaKanagawaJapan
| | - Hironori Fukuoka
- Department of Gastroenterological SurgeryYokohama City UniversityYokohamaKanagawaJapan
| | - Kentaro Miyake
- Department of Gastroenterological SurgeryYokohama City UniversityYokohamaKanagawaJapan
| | - Mayumi Ozawa
- Department of Gastroenterological SurgeryYokohama City UniversityYokohamaKanagawaJapan
| | - Takafumi Kumamoto
- Department of Surgery, Gastroenterological CenterYokohama City University Medical CenterYokohamaKanagawaJapan
| | - Ryusei Matsuyama
- Department of Gastroenterological SurgeryYokohama City UniversityYokohamaKanagawaJapan
| | - Yusuke Saigusa
- Department of BiostatisticsYokohama City UniversityYokohamaKanagawaJapan
| | - Itaru Endo
- Department of Gastroenterological SurgeryYokohama City UniversityYokohamaKanagawaJapan
| |
Collapse
|
107
|
Eslinger C, Seddighzadeh B, Yee C, Elsabbagh Z, Pai R, Hartley C, Starr J, Bekaii-Saab T, Halfdanarson TR, Sonbol MB. Clinical Outcomes and Molecular Profiling of Pancreatic Acinar Cell Carcinoma: A Retrospective Study. JCO Precis Oncol 2025; 9:e2400450. [PMID: 39772831 PMCID: PMC11706351 DOI: 10.1200/po-24-00450] [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: 07/01/2024] [Revised: 10/16/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
PURPOSE Pancreatic acinar cell carcinoma (PACC) is a rare and aggressive form of pancreatic cancer that originates in the acinar cells of the exocrine pancreas. In this study, we aimed to investigate the clinical and molecular characteristics of patients with PACC at our institution. METHODS This was a retrospective study of patients with PACC seen at Mayo Clinic between 2002 and 2023. Baseline patient characteristics, tumor pathology, treatment strategies used, and survival outcomes were analyzed. Kaplan-Meier curves were estimated using newsurv macros in SAS. RESULTS The study included a total of 65 patients with PACC. The median age at diagnosis was 66 years. Almost half of the patients (48%) presented with resectable/borderline-resectable disease (n = 28). Five-year overall survival (OS) for resectable/borderline-resectable, locally advanced/unresectable, and metastatic disease were 72.0%, 21.6%, and 20.9%, respectively. Somatic and germline next-generation sequencing identified numerous potentially actionable targets including homologous recombination (43% somatic, 33% germline), RAF alterations (29% somatic), and mismatch repair (14% somatic). CONCLUSION Our findings underscore the heterogeneity and aggressive nature of PACC. Despite the improved prognosis for patients with resectable/borderline-resectable disease, OS remains poor, particularly for those with locally advanced or metastatic disease. The identification of actionable molecular targets in a significant proportion of patients highlights the potential for personalized therapeutic approaches. Future research should focus on tailored treatment strategies to exploit these molecular vulnerabilities, which may offer new options for improving outcomes in this rare malignancy.
Collapse
Affiliation(s)
- Cody Eslinger
- Department of Hematology and Oncology, Mayo Clinic, Phoenix, AZ
| | | | - Claire Yee
- Department of Clinical Trials and Biostatistics, Mayo Clinic, Phoenix, AZ
| | - Zaid Elsabbagh
- Department of Hematology and Oncology, Mayo Clinic, Phoenix, AZ
| | - Rish Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ
| | - Chris Hartley
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Jason Starr
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, FL
| | | | | | | |
Collapse
|
108
|
Corallo C, Al-Adhami AS, Jamieson N, Valle J, Radhakrishna G, Moir J, Albazaz R. An update on pancreatic cancer imaging, staging, and use of the PACT-UK radiology template pre- and post-neoadjuvant treatment. Br J Radiol 2025; 98:13-26. [PMID: 39460945 DOI: 10.1093/bjr/tqae217] [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: 12/12/2023] [Revised: 10/01/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma continues to have a poor prognosis, although recent advances in neoadjuvant treatments (NATs) have provided some hope. Imaging assessment of suspected tumours can be challenging and requires a specific approach, with pancreas protocol CT being the primary imaging modality for staging with other modalities used as problem-solving tools to facilitate appropriate management. Imaging assessment post NAT can be particularly difficult due to a current lack of robust radiological criteria to predict response and differentiate treatment induced fibrosis/inflammation from residual tumour. This review aims to provide an update of pancreatic ductal adenocarcinoma with particular focus on three points: tumour staging pre- and post-NAT including vascular assessment, structured reporting with introduction of the PAncreatic Cancer reporting Template-UK (PACT-UK) radiology template, and the potential future role of artificial intelligence in the diagnosis and staging of pancreatic cancer.
Collapse
Affiliation(s)
- Carmelo Corallo
- Department of Radiology, St James's University Hospital, Leeds LS9 7TF, United Kingdom
| | - Abdullah S Al-Adhami
- Department of Radiology, Glasgow Royal Infirmary, Glasgow G31 2ER, United Kingdom
| | - Nigel Jamieson
- HPB Unit, Glasgow Royal Infirmary, Glasgow G31 2ER, United Kingdom
| | - Juan Valle
- Division of Cancer Sciences, University of Manchester, Manchester M20 4GJ, United Kingdom
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4 BX, United Kingdom
| | | | - John Moir
- HPB Unit, Freeman Hospital, Newcastle Upon Tyne NE7 7DN, United Kingdom
| | - Raneem Albazaz
- Department of Radiology, St James's University Hospital, Leeds LS9 7TF, United Kingdom
- University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
109
|
Kim HS, Kim W, Yun WG, Jung HS, Han Y, Lee M, Kwon W, Jang JY, Park JS. Novel Predictive Strategy Using CA19-9 and Fecal Elastase Levels to Make Treatment Decisions for Resectable Pancreatic Cancer: A Retrospective Study. Biomedicines 2024; 13:62. [PMID: 39857647 PMCID: PMC11763034 DOI: 10.3390/biomedicines13010062] [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: 11/15/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Carbohydrate antigen 19-9 (CA19-9) is used as a marker to predict recurrence and survival of patients with pancreatic ductal adenocarcinoma (PDAC). Recently, fecal elastase-1 (FE-1) has been shown to correlate with prognosis in patients with PDAC. Method: A total of 536 patients who underwent curative intent surgery between 2010 and 2019 were included in the study. The cutoff points of preoperative CA19-9 and FE-1 levels were extracted from the Youden index and previous studies. Cox proportional hazard models were used to investigate the association between preoperative tumor marker levels and survival after surgery. Results: Patients with CA19-9 ≥ 385 had more advanced T-/N-stages and lower survival rates compared to those with CA19-9 < 385. Multivariate Cox analyses demonstrated that combining preoperative tumor markers was associated with worse 3-year overall survival (both CA19-9 and FE-1 low, HR = 1.41, p = 0.044; both high, HR = 1.44, p = 0.047; CA19-9 high and FE-1 low, HR = 2.00, p < 0.001; and p for trend < 0.001). The same trend was confirmed in the analysis with recurrence-free survival. Conclusions: This study presents a new predictive strategy using combined CA19-9 and FE-1 levels to determine the treatment for resectable pancreatic cancer.
Collapse
Affiliation(s)
- Hyung Sun Kim
- Pancreatobiliary Cancer Clinic, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Woojin Kim
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Korea Medical Institute, Seoul 04522, Republic of Korea
| | - Won-Gun Yun
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hye-Sol Jung
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Youngmin Han
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Mirang Lee
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Joon Seong Park
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
110
|
Braadland PR, Farnes I, Kure EH, Yaqub S, McCann A, Ueland PM, Labori KJ, Hov JR. Indole 3-acetate and response to therapy in borderline resectable or locally advanced pancreatic cancer. Front Oncol 2024; 14:1488749. [PMID: 39759130 PMCID: PMC11695226 DOI: 10.3389/fonc.2024.1488749] [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: 08/30/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Abstract
Background/Aims It was recently reported that a higher concentration of the bacterially produced metabolite indole 3-acetate (3-IAA) in blood was linked to a better response to chemotherapy in patients with metastatic pancreatic ductal adenocarcinoma (PDAC). Here, we aimed to extend these observations to patients diagnosed with non-metastatic PDAC. Method We measured circulating 3-IAA in samples from a prospective population-based cohort of 124 patients with borderline resectable or locally advanced PDAC, collected before initiating neoadjuvant chemotherapy. The majority (61%) of the patients were treated with FOLFIRINOX. We used univariable and multivariable Cox proportional hazards regression to estimate the association between pre-treatment 3-IAA and overall survival. Results The median serum 3-IAA concentration before chemotherapy was 290 (interquartile range 203-417) ng/mL. The unadjusted hazard ratio (HR) for pre-treatment log2(3-IAA) was 0.93, 95% confidence interval (CI) [0.74-1.16], p=0.52. When adjusting for age, ECOG, CA19-9 and tumor classification, the HR for log2(3-IAA) was 0.87, 95% CI [0.68-1.12], p=0.28. Conclusion Our findings suggest that the potentiating effect of 3-IAA observed in metastatic PDAC undergoing chemotherapy may not translate to borderline resectable or locally advanced PDAC. We recommend additional clinical validation of 3-IAA's predictive value in different categories of PDAC before implementation attempts in human studies are initiated.
Collapse
Affiliation(s)
- Peder R. Braadland
- Research Institute of Internal Medicine and Norwegian PSC Research Center, Division of Surgery and Specialized Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingvild Farnes
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Oslo, Norway
| | - Elin H. Kure
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Natural Sciences and Environmental Health, University of South-Eastern Norway, Bø i Telemark, Norway
| | - Sheraz Yaqub
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Oslo, Norway
| | | | - Per M. Ueland
- BEVITAL AS, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Knut Jørgen Labori
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Oslo, Norway
| | - Johannes R. Hov
- Research Institute of Internal Medicine and Norwegian PSC Research Center, Division of Surgery and Specialized Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
111
|
Shimane G, Kitago M, Endo Y, Aiura K, Yagi H, Abe Y, Hasegawa Y, Hori S, Tanaka M, Nakano Y, Fukada J, Masugi Y, Kitagawa Y. Efficacy and safety of neoadjuvant S-1-based chemoradiotherapy in resectable and borderline-resectable pancreatic cancer: a long-term follow-up study. World J Surg Oncol 2024; 22:336. [PMID: 39707428 DOI: 10.1186/s12957-024-03609-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/01/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND/OBJECTIVES This study aimed to evaluate the safety, efficacy, and long-term outcomes of S-1-based neoadjuvant chemoradiotherapy (NACRT) in patients with resectable or borderline-resectable pancreatic ductal adenocarcinoma (PDAC). METHODS This retrospective study included patients with PDAC who underwent S-1-based NACRT at our institute between 2010 and 2017. RESULTS Forty patients were included in the study, including 15 (37.5%) with resectable PDAC and 25 (62.5%) with borderline-resectable PDAC. The NACRT completion and resection rates were 85.0% (n = 34) and 67.5% (n = 27), respectively. Several grade 3 adverse events were observed, including leukopenia (25.0%), anorexia (17.5%), neutropenia (10.0%), thrombocytopenia (7.5%), febrile neutropenia (2.5%), elevated aspartate aminotransferase/alanine aminotransferase (2.5%) levels, and hyponatremia (2.5%). The R0 resection rate was 70.4% (n = 19/27) in patients who underwent pancreatectomy. Grades 1, 2, and 3 according to the College of American Pathologists grading system were observed in 1 (3.7%), 12 (44.4%), and 14 (51.9%) patients, respectively. Over a median follow-up period of 32.9 months (interquartile range, 9.1-68.0), the 1-, 3-, and 5-year OS rates were 81.4%, 45.5%, and 30.3%, respectively, in the intention-to-treat analysis. In the curative-intent surgery cohort (n = 27), the 1-, 3-, and 5-year OS rates were 88.9%, 48.2%, and 37.0%, respectively. CONCLUSIONS S-1-based NACRT is safe and yields acceptable long-term outcomes for patients with resectable or borderline-resectable PDAC.
Collapse
Affiliation(s)
- Gaku Shimane
- Department of Surgery, Keio University School of Medicine, 35 Shinano-Machi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, 35 Shinano-Machi Shinjuku-Ku, Tokyo, 160-8582, Japan.
| | - Yutaka Endo
- Department of Surgery, Keio University School of Medicine, 35 Shinano-Machi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Koichi Aiura
- Department of Surgery, Nippon Kokan Hospital, Kanagawa, Japan
| | - Hiroshi Yagi
- Department of Surgery, Keio University School of Medicine, 35 Shinano-Machi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Yuta Abe
- Department of Surgery, Keio University School of Medicine, 35 Shinano-Machi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Yasushi Hasegawa
- Department of Surgery, Keio University School of Medicine, 35 Shinano-Machi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Shutaro Hori
- Department of Surgery, Keio University School of Medicine, 35 Shinano-Machi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Masayuki Tanaka
- Department of Surgery, Keio University School of Medicine, 35 Shinano-Machi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Yutaka Nakano
- Department of Surgery, Keio University School of Medicine, 35 Shinano-Machi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Junichi Fukada
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Yohei Masugi
- Division of Diagnostic Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, 35 Shinano-Machi Shinjuku-Ku, Tokyo, 160-8582, Japan
| |
Collapse
|
112
|
Wang J, Xu K, Zhou C, Wang X, Zuo J, Zeng C, Zhou P, Gao X, Zhang L, Wang X. A novel model based on clinical and computed tomography (CT) indices to predict the risk factors of postoperative major complications in patients undergoing pancreaticoduodenectomy. PeerJ 2024; 12:e18753. [PMID: 39713149 PMCID: PMC11663404 DOI: 10.7717/peerj.18753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 12/03/2024] [Indexed: 12/24/2024] Open
Abstract
Background Postoperative complications are prone to occur in patients after radical pancreaticoduodenectomy (PD). This study aimed to construct and validate a model for predicting postoperative major complications in patients after PD. Methods The clinical data of 360 patients who underwent PD were retrospectively collected from two centers between January 2019 and December 2023. Visceral adipose volume (VAV) and subcutaneous adipose volume (SAV) were measured using three-dimensional (3D) computed tomography (CT) reconstruction. According to the Clavien-Dindo classification system, the postoperative complications were graded. Subsequently, a predictive model was constructed based on the results of least absolute shrinkage and selection operator (LASSO) multivariate logistic regression analysis and stepwise (stepAIC) selection. The nomogram was internally validated by the training and test cohort. The discriminatory ability and clinical utility of the nomogram were evaluated by area under the receiver operating characteristic (ROC) curve (AUC), calibration curve, and decision curve analysis (DCA). Results The major complications occurred in 13.3% (n = 48) of patients after PD. The nomogram revealed that high VAV/SAV, high system inflammation response index (SIRI), high triglyceride glucose-body mass index (TyG-BMI), low prognostic nutritional index (PNI) and CA199 ≥ 37 were independent risk factors for major complications. The C-index of this model was 0.854 (95%CI [0.800-0.907]), showing excellent discrimination. The calibration curve demonstrated satisfactory concordance between nomogram predictions and actual observations. The DCA curve indicated the substantial clinical utility of the nomogram. Conclusion The model based on clinical and CT indices demonstrates good predictive performance and clinical benefit for major complications in patients undergoing PD.
Collapse
Affiliation(s)
- Jiaqi Wang
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Kangjing Xu
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Changsheng Zhou
- Department of Radiology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinbo Wang
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Junbo Zuo
- Department of General Surgery, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Chenghao Zeng
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Pinwen Zhou
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xuejin Gao
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Li Zhang
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinying Wang
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
113
|
Bhatt P, Hirsch J, Cockrum P, Kim G, Dieguez G. The Effects of Adverse Events and Associated Costs on Value-Based Care for Metastatic Pancreatic Ductal Adenocarcinoma. JOURNAL OF HEALTH ECONOMICS AND OUTCOMES RESEARCH 2024; 11:161-167. [PMID: 39717859 PMCID: PMC11664866 DOI: 10.36469/001c.124367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/03/2024] [Indexed: 12/25/2024]
Abstract
Background: Rising oncology healthcare costs have led to value-based care reimbursement models that coordinate care and improve quality while reducing overall spending. These models are increasingly important for traditional Medicare and other payers. Objectives: To compare the incidence of adverse events (AEs), AE-associated excess costs, and total cost of care (TCOC) of 3 cohorts receiving first-line treatment for metastatic pancreatic ductal adenocarcinoma (mPDAC). Methods: We conducted a retrospective analysis of administrative claims data from 2018 to 2022 using the Medicare 100% Research Identifiable Files. We examined 3 cohorts receiving mPDAC treatment: FOLFIRINOX (FFX) (oxaliplatin, irinotecan, leucovorin, 5-FU bolus and infusion); modified FFX, (5-FU infusion only); and gemcitabine/nab-paclitaxel (gem/abrax). We compared the incidence of clinically significant AEs, TCOC, components of TCOC, and costs related to AEs/treatment toxicity. Results: Patient AE rates ranged from 6.2% to 51.7%. AEs occurred more frequently in patients receiving FFX with all 4 components. Patients receiving brand name gem/abrax had lower rates of febrile neutropenia (6.2%) and neutropenia (22.2%) than those receiving FFX with no 5-FU bolus (febrile neutropenia, 9.9%; neutropenia, 36.9%) and FFX with all 4 components (febrile neutropenia, 6.9%; neutropenia, 30.4%). Rates of most nonhematologic AEs were higher in patients receiving FFX with all 4 components, with diarrhea occurring in 28.3%, abdominal pain in 31.5%, and nausea/vomiting in 41.5% of patients. TCOC was lower in the gem/abrax cohort: 6505 v s F F X w i t h n o 5 - F U b o l u s ( 6995) and FFX with all 4 components ( 7142 ) p e r a d m i n i s t r a t i o n . T h e d e v e l o p m e n t o f a n y s t u d i e d h e m a t o l o g i c A E w a s a s s o c i a t e d w i t h a m e a n e x c e s s c o s t o f 5993 per administration, while the development of any studied nonhematological AE was associated with a mean per-administration excess cost of $3665. Discussion: Treatment decisions intended to minimize chemotherapy costs may lead to suboptimal decisions if the goal is to reduce TCOC. Our research suggests FFX is more costly than gem/abrax (TCOC per administration). Patients receiving gem/abrax were older and had higher baseline Charlson Comorbidity Index scores; however, other factors may be important in driving cost differences. Conclusions: Irrespective of drug cost, chemotherapy leading to a significant increase in AEs is associated with higher TCOC.
Collapse
|
114
|
Yang H, Yu P, Gong J. Prognostic biomarker MICAL2 and associates with proliferation, migration and immune infiltration in pancreatic adenocarcinoma. J Appl Genet 2024:10.1007/s13353-024-00919-3. [PMID: 39661267 DOI: 10.1007/s13353-024-00919-3] [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: 12/07/2023] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 12/12/2024]
Abstract
To elucidate the crucial function of MICAL2 as a potential immunotherapeutic target and a predictive biomarker in PAAD. The expression of MICAL2 in pan-cancer was investigated using public database, and the expression of MICAL2 in PAAD was validated using tissue samples. The diagnostic and prognostic significance of MICAL2 in PAAD was assessed through the application of ROC curves and Kaplan-Meier curves. The correlation between MICAL2 and infiltrating immune cells and immune checkpoints in PAAD was researched using the TIMER and TCGA databases. In vitro studies involved the evaluation of the biological functions of MICAL2 in human PAAD cells through the knockdown of MICAL2 expression using shRNA. Compared to corresponding normal tissues, the expression of MICAL2 exhibits significant differences in various cancers. Specifically, the level of MICAL2 expression is significantly increased in PAAD. Moreover, MICAL2 demonstrates considerable diagnostic potential in PAAD patients, and its elevated expression is indicative of an unfavorable prognosis. The differential expression of MICAL2 is related to infiltrating immune cells, immune cell markers, and immune checkpoints in PAAD. In ASPC-1 and PANC-1 cells, when MICAL2 was knocked down, there was a notable suppression of proliferation, migration, and invasion. MICAL2 functions as a significant predictor and promising immunotherapeutic target for prognosis assessment in PAAD. It has a pivotal function in fostering the growth and migration of PAAD cells.
Collapse
Affiliation(s)
- Huachao Yang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Department of Breast, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China
| | - Pingping Yu
- Department of Health Management, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Jianping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
115
|
Qin D, Xi P, Huang K, Jiang L, Yao Z, Wei R, Li S. Nomogram for predicting post-progression-free survival in patients with recurrent pancreatic ductal adenocarcinoma after radical surgery: a retrospective analysis. Front Med (Lausanne) 2024; 11:1486750. [PMID: 39712186 PMCID: PMC11659012 DOI: 10.3389/fmed.2024.1486750] [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: 08/26/2024] [Accepted: 11/20/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Radical resection is the only curative method for patients with pancreatic adenocarcinoma (PDAC). However, nearly 85% of PDAC patients suffer from local or distant recurrence within 5 years after curative resection. The progression of recurrent lesions accelerates the mortality rate in PDAC patients. However, the influence of clinicopathological factors on post-progression-free survival (PPFS), defined as the period from tumor recurrence to the timing of the progression of recurrent lesions, has rarely been discussed. The present study aimed to explore the independent prognostic factors for PPFS and construct a nomogram for PPFS prediction. MATERIALS AND METHODS The 200 recurrent PDAC patients were divided into training and validation groups by leave-one-out cross-validation. The patients' clinicopathological characteristics were compared through a chi-square test. Meanwhile, these factors were enrolled in the univariate and multivariate COX regression to find the independent prognostic factors of PPFS. Moreover, the Kaplan-Meier survival analysis based on the independent prognostic factors was performed. Finally, we constructed a nomogram model for PPFS prediction, followed by an effectiveness examination. RESULTS PDAC patients who received multi-agent chemotherapy after surgery showed a longer PPFS than the single-agent chemotherapy group. PDAC patients who received multi-agent chemotherapy after recurrence showed a similar PPFS compared to the single-agent chemotherapy group. Local recurrence with distant metastases, early recurrence, lympho-vascular invasion, higher T stage, and higher N stage predicted shorter PPFS in recurrent PDAC patients. Finally, a nomogram to indicate the progression of recurrent lesions was constructed. CONCLUSION Multi-agent chemotherapy is recommended for PDAC patients after surgery. Meanwhile, single-agent chemotherapy also deserves consideration after tumor recurrence. Moreover, the nomogram could be used in PPFS prediction.
Collapse
Affiliation(s)
| | | | | | | | | | - Ran Wei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shengping Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
116
|
Lin Y, Liu T, Hu Y, Xu Y, Wang J, Guo S, Xie S, Sun H. Assessment of vascular invasion of pancreatic ductal adenocarcinoma based on CE-boost black blood CT technique. Insights Imaging 2024; 15:293. [PMID: 39636361 PMCID: PMC11621291 DOI: 10.1186/s13244-024-01870-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: 06/27/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
OBJECTIVES To explore the diagnostic efficacy of advanced intelligent clear-IQ engine (AiCE) and adaptive iterative dose reduction 3D (AIDR 3D), combination with and without the black blood CT technique (BBCT), for detecting vascular invasion in patients diagnosed with nonmetastatic pancreatic ductal adenocarcinoma (PDAC). METHODS A total of 35 consecutive patients diagnosed with PDAC, proceeding with contrast-enhanced abdominal CT scans, were enrolled in this study. The arterial and portal venous phase images were reconstructed using AiCE and AIDR 3D. The corresponding BBCT images were established as AiCE-BBCT and AIDR 3D-BBCT, respectively. Two observers scored the image quality independently. Cohen's kappa (k) value or intraclass correlation coefficient (ICC) was used to analyze consistency. The diagnostic performance of four algorithms in detecting vascular invasion in PDAC patients was assessed using the area under the curve (AUC). RESULTS The AiCE and AiCE-BBCT groups demonstrated superior image noise and diagnostic acceptability compared with AIDR 3D and AIDR 3D-BBCT groups (all p < 0.001), and the k value was 0.861-0.967 for both reviewers. In terms of diagnostic capability for vascular invasion in PDAC, the AiCE-BBCT group exhibited higher specificity (95.0%) and sensitivity (93.3%) compared to the AIDR 3D and AIDR 3D-BBCT groups, with an AUC of 0.942 (95% CI: 0.849-1.000, p < 0.05). Furthermore, all vascular evaluations conducted using AiCE-BBCT demonstrated better consistency (ICC: 0.847-0.935). CONCLUSION The BBCT technique in conjunction with AiCE could lead to notable enhancements in both the image quality of PDAC images and the diagnostic performance for tumor vascular invasion. CRITICAL RELEVANCE STATEMENT Better diagnostic accuracy of vascular invasion of PDAC based on BBCT in combination with an AiCE is a critical factor in determining treatment strategies and patient outcomes. KEY POINTS Identifying vascular invasion of PDAC is important for prognostication. Combined images provide improved image quality and higher diagnostic accuracy. Combined images can excellently display the vascular wall and invasion.
Collapse
Affiliation(s)
- Yue Lin
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Tongxi Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Yingying Hu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Yinghao Xu
- CT Clinical Research Department, CT Business Unit, Canon Medical Systems (China) Co., Ltd., Beijing, China
| | - Jian Wang
- CT Clinical Research Department, CT Business Unit, Canon Medical Systems (China) Co., Ltd., Beijing, China
| | - Sijia Guo
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Sheng Xie
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Hongliang Sun
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
117
|
Faccenda V, Panizza D, Niespolo RM, Colciago RR, Rossano G, De Sanctis L, Gandola D, Ippolito D, Arcangeli S, De Ponti E. Synchronized Contrast-Enhanced 4DCT Simulation for Target Volume Delineation in Abdominal SBRT. Cancers (Basel) 2024; 16:4066. [PMID: 39682252 DOI: 10.3390/cancers16234066] [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: 10/24/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: To present the technical aspects of contrast-enhanced 4DCT (ce4DCT) simulation for abdominal SBRT. Methods: Twenty-two patients underwent two sequential 4DCT scans: one baseline and one contrast-enhanced with personalized delay time (tdelay) calculated to capture the tumor in the desired contrast phase, based on diagnostic triple-phase CT. The internal target volume (ITV) was delineated on ten contrast phases, and a panel of three experts qualitatively evaluated tumor visibility. Aortic HU values were measured to refine the tdelay for subsequent patients. The commonly used approach of combining triple-phase CT with unenhanced 4DCT was simulated, and differences in target delineation were evaluated by volume, centroid shift, Dice and Jaccard indices, and mean distance agreement (MDA). The margins required to account for motion were calculated. Results: The ce4DCT acquisitions substantially improved tumor visibility over the entire breathing cycle in 20 patients, according to the experts' unanimous evaluation. The median contrast peak time was 54.5 s, and the washout plateau was observed at 70.3 s, with mean peak and plateau HU values of 292 ± 59 and 169 ± 25. The volumes from the commonly used procedure (ITV2) were significantly smaller than the ce4DCT volumes (ITV1) (p = 0.045). The median centroid shift was 4.7 mm. The ITV1-ITV2 overlap was 69% (Dice index), 53% (Jaccard index), and 2.89 mm (MDA), with the liver volumes showing significantly lower indices compared to the pancreatic volumes (p ≤ 0.011). The margins required to better encompass ITV1 were highly variable, with mean values ≥ 4 mm in all directions except for the left-right axis. Conclusions: The ce4DCT simulation was feasible, resulting in optimal tumor enhancement with minimal resource investment, while significantly mitigating uncertainties in SBRT planning by addressing poor visibility and respiratory motion. Triple-phase 3DCT with unenhanced 4DCT led to high variability in target delineation, making the isotropic margins ineffective.
Collapse
Affiliation(s)
- Valeria Faccenda
- Medical Physics Department, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Denis Panizza
- Medical Physics Department, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milan Bicocca, 20126 Milan, Italy
| | - Rita Marina Niespolo
- Radiation Oncology Department, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | | | - Giulia Rossano
- School of Medicine and Surgery, University of Milan Bicocca, 20126 Milan, Italy
- Radiation Oncology Department, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Lorenzo De Sanctis
- School of Medicine and Surgery, University of Milan Bicocca, 20126 Milan, Italy
- Radiation Oncology Department, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Davide Gandola
- Diagnostic Radiology Department, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Davide Ippolito
- School of Medicine and Surgery, University of Milan Bicocca, 20126 Milan, Italy
- Diagnostic Radiology Department, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Stefano Arcangeli
- School of Medicine and Surgery, University of Milan Bicocca, 20126 Milan, Italy
- Radiation Oncology Department, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Elena De Ponti
- Medical Physics Department, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milan Bicocca, 20126 Milan, Italy
| |
Collapse
|
118
|
Kohara Y, Yasuda S, Nagai M, Nakamura K, Matsuo Y, Terai T, Doi S, Sakata T, Sho M. Prognostic significance of creatine kinase in resected pancreatic cancer. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2024; 31:906-916. [PMID: 39489619 DOI: 10.1002/jhbp.12081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
BACKGROUND Creatine kinase (CK) levels decrease with cancer progression and muscle wasting, but its association with pancreatic ductal adenocarcinoma (PDAC) remains unclear. The aim of this study was to investigate CK as a prognostic biomarker and surrogate marker for muscle mass in patients with PDAC. METHODS A retrospective analysis of 476 patients with PDAC was conducted. CK levels were categorized into low and high groups using receiver-operating characteristic (ROC) curve analysis. RESULTS Among the 476 patients, 200 (42.0%) and 276 (58.0%) were classified into the low and high CK groups, respectively. The low CK group had significantly poorer overall survival (p < .001) and recurrence-free survival (p < .001) compared to the high CK group. Multivariate analysis identified low CK as an independent poor prognostic factor (p < .001). The low CK group had significantly lower skeletal muscle index (p = .048) than the high CK group; however, the difference was slight and not significantly associated with sarcopenia. Additionally, combined risk assessment incorporating CK and resectability facilitated a more nuanced prognostic stratification. CONCLUSIONS CK served as a reliable prognostic marker independent from resectability but was less effective as a marker for sarcopenia in PDAC.
Collapse
Affiliation(s)
| | - Satoshi Yasuda
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Minako Nagai
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Kota Nakamura
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Yasuko Matsuo
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Taichi Terai
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Shunsuke Doi
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Takeshi Sakata
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Masayuki Sho
- Department of Surgery, Nara Medical University, Nara, Japan
| |
Collapse
|
119
|
Padillo-Ruiz J, Garcia C, Suarez G, Blanco G, Muñoz-Bellvis L, Justo I, García-Domingo MI, Ausania F, Muñoz-Forner E, Serrablo A, Martin E, Díez L, Cepeda C, Marin L, Alamo J, Bernal C, Pereira S, Calero F, Laga I, Paterna S, Cugat E, Fondevila C, López-Guerra D, Gallego-Jiménez I, Borrero-Martín JJ, Gomez-Bravo MÁ, Tinoco J, Sabater L. Intraoperative liquid biopsy as a tool for detecting R1 resection during pancreatoduodenectomy in patients with pancreatic carcinoma: the CETUPANC trial (part II). Int J Surg 2024; 110:7798-7805. [PMID: 39806742 PMCID: PMC11634100 DOI: 10.1097/js9.0000000000002153] [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: 09/18/2024] [Accepted: 11/07/2024] [Indexed: 01/16/2025]
Abstract
INTRODUCTION A positive surgical margin (R1 resection) is a relevant risk factor for local recurrence in patients with pancreatic ductal adenocarcinoma of the pancreas (PDAC). An intraoperative liquid biopsy (ILB) based on tumor cell mobilization could help to detect R1 resection intraoperatively. OBJECTIVE To evaluate the potential role of the intraoperative circulating tumor cells (CTCs) and cluster mobilization on the R0/R1 detection. METHODS Sixty-three patients with resectable PDAC of the head of the pancreas were prospective enrolled under the CETUPANC trial. Open pancreaticoduodenectomy (PD) was done in all patients. Intraoperative CTCs and clusters were determined during PD. RESULTS The overall rate of R1 resection was 34.9% (22/63 patients). Multivariate analysis showed that factors associated with R1 resection (AUC=0.920) were the presence of undifferentiated G3 tumor (P=0.017), microscopic vascular invasion (P=0.016), and the intraoperative increase of both free CTCs and clusters in portal vein determination from the beginning to the end of the surgery (P=0.002 and P=0.005, respectively). A specific logistic regression model, including delta end to baseline CTCs and cluster mobilization to achieve a combined cut-off to detect R1 detection was calculated (AUC=0.799). The obtained R1-index based on ILB had 84% of sensitivity and 68% of specificity to detect R1 resection. CONCLUSIONS The ILB based on the intraoperative mobilization of CTCs and clusters from the beginning to the end of the PD was a predictive factor to detect R1 resection in patients with PDAC.
Collapse
Affiliation(s)
| | | | | | - Gerardo Blanco
- Badajoz University Hospital, University of Extremadura, Badajoz
| | - Luis Muñoz-Bellvis
- University Hospital of Salamanca, Salamanca Biosanitary Institute, University of Salamanca, Salamanca
| | - Iago Justo
- University Hospital October 12 in Madrid, Madrid
| | | | - Fabio Ausania
- Hospital-Clinic, August Pi i Sunyer Biomedical Research Institute, University of Barcelona, Barcelona
| | - Elena Muñoz-Forner
- Valencia Clinical Hospital, University of Valencia, Biomedical Research Institute, Incliva, Valencia
| | | | | | | | | | - Luis Marin
- Virgen del Rocío University Hospital, IBIS, Seville
| | - Jose Alamo
- Virgen del Rocío University Hospital, IBIS, Seville
| | | | | | | | - Imán Laga
- Virgen del Rocío University Hospital, IBIS, Seville
| | | | | | - Constantino Fondevila
- Hospital-Clinic, August Pi i Sunyer Biomedical Research Institute, University of Barcelona, Barcelona
| | | | | | | | | | - Jose Tinoco
- Virgen del Rocío University Hospital, IBIS, Seville
| | - Luis Sabater
- Valencia Clinical Hospital, University of Valencia, Biomedical Research Institute, Incliva, Valencia
| |
Collapse
|
120
|
Thomas AS, Tehranifar P, Kwon W, Shridhar N, Sugahara KN, Schrope BA, Chabot JA, Manji GA, Genkinger JM, Kluger MD. Trends in the Care of Locally Advanced Pancreatic Cancer in the Modern Era of Chemotherapy. J Surg Oncol 2024; 130:1589-1604. [PMID: 39348434 DOI: 10.1002/jso.27851] [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: 04/02/2024] [Revised: 08/04/2024] [Accepted: 08/18/2024] [Indexed: 10/02/2024]
Abstract
INTRODUCTION Current guidelines for treatment for locally advanced pancreatic cancer recommend chemotherapy ± radiation, or radiation alone when multimodal therapy is contraindicated. In a subset of patients, guideline-recommended treatment (GRT) achieves sufficient response to qualify for potentially curative resection. This study evaluated trends in treatment utilization and aimed to identify barriers to GRT. METHODS Patients with clinical T4M0 disease in the National Cancer Database from 2010 to 2017 were included. Potential predictors were assessed by relative risk regression with Poisson distribution and compared by log-link function. RESULTS In total, 28 056 patients met the criteria. Among 17 059 (67.67%) patients treated primarily with chemotherapy, 41.19% also had radiation and 8.89% went onto resection. Many received no cancer-directed treatment or failed to receive GRT. Another 710 patients had radiation (±surgery) without chemotherapy despite few contraindications to chemotherapy. Over time, patients were more likely to undergo resection after chemotherapy (aRR = 1.58; p < 0.0001) and less likely to have chemoradiation (aRR = 0.78; p < 0.0001) or go untreated (aRR = 0.90; p < 0.0001). Socioeconomic factors (race, education, income, and insurance status) affected the likelihood of receiving chemotherapy and surgery. Median overall survival (OS) was significantly improved for patients treated with chemotherapy and particularly in those patients who went on to receive RT or undergo surgical resection. OS was also longer for patients treated at high-volume academic centers. Patients insured by Medicaid, Medicare, or those without insurance had worse OS. CONCLUSIONS Despite improvement over time, many patients go untreated. Clinical factors were influential, but the impact of vulnerable social standing suggests persistent inequity in access to care.
Collapse
Affiliation(s)
- Alexander S Thomas
- Department of Surgery, Division of Gastrointestinal and Endocrine Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - Parisa Tehranifar
- Herbert Irving Comprehensive Cancer Center Cancer Population Science Program, New York, New York, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Wooil Kwon
- Department of Surgery, Division of Gastrointestinal and Endocrine Surgery, Columbia University Irving Medical Center, New York, New York, USA
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Nupur Shridhar
- Department of Surgery, Division of Gastrointestinal and Endocrine Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - Kazuki N Sugahara
- Department of Surgery, Division of Gastrointestinal and Endocrine Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - Beth A Schrope
- Department of Surgery, Division of Gastrointestinal and Endocrine Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - John A Chabot
- Department of Surgery, Division of Gastrointestinal and Endocrine Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - Gulam A Manji
- Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, New York, New York, USA
| | - Jeanine M Genkinger
- Herbert Irving Comprehensive Cancer Center Cancer Population Science Program, New York, New York, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Michael D Kluger
- Department of Surgery, Division of Gastrointestinal and Endocrine Surgery, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
121
|
Kang M, Yoo S, Jung Y, Lim H, Lee MH, Ryu JK, Lee JI. Factors affecting peripheral neuropathy induced by nanoparticle albumin-bound paclitaxel in patients with pancreatic cancer. Br J Clin Pharmacol 2024; 90:3232-3241. [PMID: 39152547 PMCID: PMC11602956 DOI: 10.1111/bcp.16210] [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: 05/22/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/19/2024] Open
Abstract
AIM Chemotherapy-induced peripheral neuropathy (CIPN) is a major toxicity limiting the use of nab-paclitaxel (Nab-P) in treating patients with pancreatic cancer. The aim of this study was to identify the factors affecting CIPN using patient-reported outcome measures and the minimally invasive volumetric absorptive microsampling (VAMS) technique. METHODS The maximum concentrations of paclitaxel (Cmax) were measured from 81 VAMS samples collected from 44 participants with pancreatic cancer. The association between CIPN development and demographic, clinical and pharmacokinetic factors was determined using univariable and multivariable logistic regression. The association between CIPN severity and the factors was evaluated using Spearman's rank correlation. The impact of Cmax and the number of treatment cycles on the severity was assessed using multivariable linear regression. RESULTS The development of CIPN was significantly associated with cumulative dose (odds ratio 1.005, 95% confidence interval [CI] 1.003-1.007), treatment cycles (3.47, 2.25-5.85), alkaline phosphate (0.992, 0.985-0.998) and age (1.092, 1.020-1.179), with an area under the receiver operating characteristic curve of 0.89 (95% CI 0.83-0.95). The severity of CIPN significantly worsened with increasing cumulative dose (coefficient 0.58, 95% CI 0.44-0.69), treatment cycles (0.57, 0.44-0.68) and age (0.18, 0.00-0.35). The severity of CIPN was predictable from treatment cycles (P = .0002) and Cmax (P = .01). CONCLUSION The higher the cumulative dose of Nab-P, treatment cycles and age, the more frequently and severely do the patients experience CIPN. In predicting the severity of CIPN using Cmax, minimally invasive VAMS is a feasible alternative to venous blood sampling.
Collapse
Affiliation(s)
- Minseo Kang
- College of Pharmacy and Research Institute of Pharmaceutical SciencesSeoul National UniversitySeoulRepublic of Korea
| | - Seunghyun Yoo
- College of Pharmacy and Research Institute of Pharmaceutical SciencesSeoul National UniversitySeoulRepublic of Korea
| | - Yeolmae Jung
- College of Pharmacy and Research Institute of Pharmaceutical SciencesSeoul National UniversitySeoulRepublic of Korea
- Present address:
Department of Medical Information and Medical ScienceBoryung Inc.SeoulRepublic of Korea
| | - Hayun Lim
- College of Pharmacy and Research Institute of Pharmaceutical SciencesSeoul National UniversitySeoulRepublic of Korea
| | - Myeong Hwan Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University HospitalSeoul National University College of MedicineSeoulRepublic of Korea
| | - Ji Kon Ryu
- Department of Internal Medicine and Liver Research Institute, Seoul National University HospitalSeoul National University College of MedicineSeoulRepublic of Korea
| | - Jangik I. Lee
- College of Pharmacy and Research Institute of Pharmaceutical SciencesSeoul National UniversitySeoulRepublic of Korea
| |
Collapse
|
122
|
Ingaldi C, D’Ambra V, Ricci C, Alberici L, Minghetti M, Grego D, Cavallaro V, Casadei R. Clinicopathological predictive factors in long-term survivors who underwent surgery for pancreatic ductal adenocarcinoma: A single-center propensity score matched analysis. World J Surg 2024; 48:3001-3013. [PMID: 39542862 PMCID: PMC11619735 DOI: 10.1002/wjs.12397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/06/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Long-term survivors (LTSs) after pancreatic resection of pancreatic ductal adenocarcinoma (PDAC) represent a particular subgroup of patients that remains poorly understood. The primary endpoint was to identify clinicopathological factors associated with LTSs after pancreatic resection for PDAC. METHODS This was a retrospective study of patients who had undergone pancreatic resection for PDAC. Long survival was defined as a patient who survived at least 60 months. Patients were divided in two groups: LTS and short-term survivor (STS). The two groups were compared regarding epidemiological, clinical, and pathological data. Propensity score matching (PSM) was used to reduce selection bias with a 1:2 ratio. Multivariable analysis of significative predictive factors before and after PSM was done. RESULTS Three hundred and thirty-three patients were enrolled: 46 (13.8%) in the LTS group and 287 (86.2%) in the STS group. Using PSM, 138 patients were analyzed: 46 in the LTS group and 92 in the STS group. At the multivariate analysis of significative predictive factor after PSM, adjuvant chemotherapy, well-differentiated tumors (G1), and R0 status were related to long-term survival (p = 0.052, 0.010 and p = 0.019, respectively). Kaplan-Meier survival curves confirmed these data. Additionally, Kaplan-Meier survival curves showed that pathological stage I was a favorable factor with respect to stage II, III, and IV. CONCLUSIONS Long-term survival is possible after pancreatic cancer resection, even if in a small percentage. Significant predictors of long-term survival are administration of adjuvant chemotherapy, American Join Committee on Cancer stage I, well-differentiated tumor (G1), and R0 resection.
Collapse
Affiliation(s)
- Carlo Ingaldi
- Division of Pancreatic SurgeryIRCCSAzienda Ospedaliero Universitaria di BolognaBolognaItaly
- Department of Internal Medicine and Surgery (DIMEC)Alma Mater StudiorumUniversity of BolognaS.Orsola‐Malpighi HospitalBolognaItaly
| | - Vincenzo D’Ambra
- Division of Pancreatic SurgeryIRCCSAzienda Ospedaliero Universitaria di BolognaBolognaItaly
- Department of Internal Medicine and Surgery (DIMEC)Alma Mater StudiorumUniversity of BolognaS.Orsola‐Malpighi HospitalBolognaItaly
| | - Claudio Ricci
- Division of Pancreatic SurgeryIRCCSAzienda Ospedaliero Universitaria di BolognaBolognaItaly
- Department of Internal Medicine and Surgery (DIMEC)Alma Mater StudiorumUniversity of BolognaS.Orsola‐Malpighi HospitalBolognaItaly
| | - Laura Alberici
- Division of Pancreatic SurgeryIRCCSAzienda Ospedaliero Universitaria di BolognaBolognaItaly
- Department of Internal Medicine and Surgery (DIMEC)Alma Mater StudiorumUniversity of BolognaS.Orsola‐Malpighi HospitalBolognaItaly
| | - Margherita Minghetti
- Division of Pancreatic SurgeryIRCCSAzienda Ospedaliero Universitaria di BolognaBolognaItaly
- Department of Internal Medicine and Surgery (DIMEC)Alma Mater StudiorumUniversity of BolognaS.Orsola‐Malpighi HospitalBolognaItaly
| | - Davide Grego
- Division of Pancreatic SurgeryIRCCSAzienda Ospedaliero Universitaria di BolognaBolognaItaly
- Department of Internal Medicine and Surgery (DIMEC)Alma Mater StudiorumUniversity of BolognaS.Orsola‐Malpighi HospitalBolognaItaly
| | - Virginia Cavallaro
- Division of Pancreatic SurgeryIRCCSAzienda Ospedaliero Universitaria di BolognaBolognaItaly
- Department of Internal Medicine and Surgery (DIMEC)Alma Mater StudiorumUniversity of BolognaS.Orsola‐Malpighi HospitalBolognaItaly
| | - Riccardo Casadei
- Division of Pancreatic SurgeryIRCCSAzienda Ospedaliero Universitaria di BolognaBolognaItaly
- Department of Internal Medicine and Surgery (DIMEC)Alma Mater StudiorumUniversity of BolognaS.Orsola‐Malpighi HospitalBolognaItaly
| |
Collapse
|
123
|
Zhou J, Dong G, Jing X, Huang G, Wang Z, Peng M, Zhou Y, Yu X, Yu J, Han Z, Liu F, Gao H, Zhang Y, Cheng Z, Ye X, Liang P. Image-guided percutaneous microwave ablation for unresectable pancreatic cancers: A multicenter retrospective study. Eur J Radiol 2024; 181:111720. [PMID: 39326234 DOI: 10.1016/j.ejrad.2024.111720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/22/2024] [Accepted: 09/02/2024] [Indexed: 09/28/2024]
Abstract
OBJECTIVE This study aims to assess the feasibility, effectiveness, and safety of image-guided percutaneous microwave ablation (PMWA) for unresectable pancreatic cancer. METHODS In this retrospective study, 72 patients from four hospitals were enrolled between November 2009 and October 2022. Descriptive statistics were employed to describe the patients' characteristics and prognostic factors. The primary endpoint compassed the complete ablation rate (CAR), incidence of complications and the pain relief rate (PRR). RESULTS The median age of the 72 patients was 61 (interquartile range (IQR) 52.5-67.0) years, with 62.5 % (45/72) being male. 26 cases received computed tomography (CT) guidance; 46 cases received ultrasound guidance. A total of 74 tumors were identified (2 in 2 patients), with 56.8 % (42/74) at the body and tail, and the rest at the head and neck. Overall, 73 ablation sessions were carried out, achieving a technical success rate (TSR) of 100 %. The CAR was 40.5 % (30/74). The median follow-up time was 4.6 (1-43.4) months. 50 % (36/72) of patients had died with a median overall survival (OS) of 5.6 (1-27) months. Regarding complications, 18.1 % (13/72) of cases were classified as grade I and II, and 9.8 % (7/72) as grade IIIa. Before surgery, 33 patients experienced pain symptoms, and the postoperative PRR was 96.7 % (32/33). The average pain score decreased from 6.3 (4-10) before surgery to 2.0 (0-8) after ablation (P<0.001). CONCLUSIONS Image-guided PMWA for unresectable pancreatic cancer is safe and feasible, effectively relieving cancer pain and improving patients' the quality of life.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China 100853; Chinese PLA Medical School, Beijing, China
| | - Gang Dong
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China 450000
| | - Xiang Jing
- Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China
| | - Guanghui Huang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China 250021
| | - Zhen Wang
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China 100853
| | - Mengfan Peng
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China 450000
| | - Yan Zhou
- Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China
| | - Xiaoling Yu
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China 100853
| | - Jie Yu
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China 100853
| | - Zhiyu Han
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China 100853
| | - Fangyi Liu
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China 100853
| | - Hongjian Gao
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing 100124, China
| | - Yubo Zhang
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing 100124, China
| | - Zhigang Cheng
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China 100853.
| | - Xin Ye
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China 250014.
| | - Ping Liang
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China 100853.
| |
Collapse
|
124
|
Tang R, Liu M, Shu Q, Chen X, Cai L. Performance of fibroblast activating protein inhibitor PET imaging for pancreatic neoplasms assessment: a systematic review and meta-analysis. Eur Radiol 2024; 34:7804-7812. [PMID: 38907099 DOI: 10.1007/s00330-024-10843-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/05/2024] [Accepted: 04/21/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Recent studies have shown the potential of fibroblast activating protein inhibitor (FAPI) PET imaging for pancreatic cancer assessment. PURPOSE This article is dedicated to comparing the diagnostic efficacy of FAPI PET and [18F]fluorodeoxyglucose (FDG) PET in the evaluation of primary tumors, lymph nodes, and distant metastases in pancreatic cancer. METHODS In this review, we conducted a systematic search of studies published in PubMed and Web of Science databases up to September 18, 2023. All included studies used radionuclide labeled FAPI and FDG as PET diagnostic tracers to evaluate their applicability in patients with pancreatic cancer. RESULTS The FAPI PET imaging group showed significantly higher sensitivity in the detection of primary lesions (1.000, [95% CI: 0.999-1.000]), lymph node metastases (0.624 [95% CI: 0.391-0.834]) and distant metastatic (0.965 [95% CI: 0.804-1.000]) in pancreatic cancer compared to the FDG PET imaging group (0.889 [95% CI: 0.788-0.966], 0.373 [95% CI: 0.163-0.606] and 0.889 [95% CI: 0.689-0.999], respectively). Furthermore, the maximum standardized uptake value (SUVmax) in FAPI PET imaging is significantly higher than that in FDG imaging for primary lesions (mean difference (MD) = 7.51, 95% CI: 5.34-9.67). CONCLUSION Compared with [18F]FDG PET/CT, FAPI PET imaging showed higher sensitivity, SUVmax. This method can be effectively utilized for the evaluation of pancreatic cancer. CLINICAL RELEVANCE STATEMENT Fibroblast activating protein inhibitor PET may be a better alternative to [18F]FDG in evaluating primary pancreatic cancer, lymph node metastases, and distant metastases. KEY POINTS Fibroblast activating protein inhibitor (FAPI) PET is compared with FDG PET for evaluating pancreatic cancer. Multiple radiolabeled FAPI variants have shown promising results in the diagnosis of pancreatic cancer. FAPI PET imaging effectively helps clinicians diagnose and stage pancreatic cancer.
Collapse
Affiliation(s)
- Ranbie Tang
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, 646000, Luzhou, Sichuan, PR China
- Institute of Nuclear Medicine, Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
| | - Mengna Liu
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, 646000, Luzhou, Sichuan, PR China
- Institute of Nuclear Medicine, Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
| | - Qiaoqiao Shu
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, 646000, Luzhou, Sichuan, PR China
- Institute of Nuclear Medicine, Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
| | - Xi Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, 646000, Luzhou, Sichuan, PR China
- Institute of Nuclear Medicine, Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
| | - Liang Cai
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China.
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, 646000, Luzhou, Sichuan, PR China.
- Institute of Nuclear Medicine, Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China.
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, 400010, Chongqing, PR China.
| |
Collapse
|
125
|
Yun WG, Gil J, Choi H, Han Y, Jung HS, Cho YJ, Suh M, Kwon W, Lee YS, Cheon GJ, Jang JY. Prospective Comparison of [ 18F]FDG and [ 18F]AIF-FAPI-74 PET/CT in the Evaluation of Potentially Resectable Pancreatic Ductal Adenocarcinoma. Mol Imaging Biol 2024; 26:1068-1077. [PMID: 39365411 PMCID: PMC11634952 DOI: 10.1007/s11307-024-01950-w] [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: 03/21/2024] [Revised: 07/29/2024] [Accepted: 08/24/2024] [Indexed: 10/05/2024]
Abstract
PURPOSE Accurate clinical staging of potentially resectable pancreatic ductal adenocarcinoma (PDAC) is critical for establishing optimal treatment strategies. While the efficacy of fluorine-18-fluorodeoxyglucose ([18F]FDG) positron emission tomography/computed tomography (PET/CT) in clinical staging is unclear, PET/CT detecting fibroblast-activation protein (FAP) expression has recently received considerable attention for detecting various tumors, including PDAC, with high sensitivity. We explored the efficacy of [18F]FDG and [18F]AIF-FAPI-74 PET/CT in the initial evaluation of potentially resectable PDAC. PROCEDURES Between 2021 and 2022, twenty participants with newly diagnosed potentially resectable PDAC were enrolled. After the initial evaluation with pancreatic CT, [18F]FDG PET/CT, and [18F]AIF-FAPI-74 PET/CT, treatment strategies were determined considering the participant's general status, clinical staging, and resectability. Pathological information from the surgical specimens was only available in 17 participants who underwent curative-intent surgery. Head-to-head comparisons of quantitative radiotracer uptake and diagnostic performance were performed among imaging modalities. RESULTS [18F]AIF-FAPI-74 PET/CT showed a significantly higher maximum standardized uptake value than [18F]FDG PET/CT did in evaluating primary pancreatic lesions (median [interquartile range]; 12.6 [10.7-13.7] vs. 6.3 [4.8-9.2]; P < 0.001). In contrast, [18F]AIF-FAPI-74 PET/CT showed a significantly lower mean standardized uptake value than [18F]FDG PET/CT did in evaluating background organ (median [interquartile range]) 0.8 [0.7-0.9] vs. 2.6 [2.3-2.7]; P < 0.001). In addition, the sensitivity of [18F]AIF-FAPI-74 PET/CT in detecting metastatic lymph nodes was higher than that of [18F]FDG PET/CT (50.0% vs. 0.0%; P = 0.026). CONCLUSION This study demonstrated that [18F]AIF-FAPI-74 PET/CT could improve the clinical staging of potentially resectable PDAC.
Collapse
Affiliation(s)
- Won-Gun Yun
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Joonhyung Gil
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno- gu, Seoul, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno- gu, Seoul, Republic of Korea
- Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Youngmin Han
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Hye-Sol Jung
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Young Jae Cho
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Minseok Suh
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno- gu, Seoul, Republic of Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno- gu, Seoul, Republic of Korea
- Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
- Cancer Research Institute & Institute on Aging, Seoul National University, Seoul, Republic of Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| |
Collapse
|
126
|
Cheng K, Li X, Lv W, Zhao G, Zhou R, Chang C, Yang H, Li R, Li Z, Chen Y, Yi C, Yan O, Xiao C, Zhang Y, Xiong J, Huang Z, Shao W, You X, Guo W, He D, Ling W, Wang R, Tian B, Zhao C, Cao D. Spatial interactions of immune cells as potential predictors to efficacy of toripalimab plus chemotherapy in locally advanced or metastatic pancreatic ductal adenocarcinoma: a phase Ib/II trial. Signal Transduct Target Ther 2024; 9:321. [PMID: 39582060 PMCID: PMC11586424 DOI: 10.1038/s41392-024-02031-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/10/2024] [Accepted: 10/23/2024] [Indexed: 11/26/2024] Open
Abstract
Advanced pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis. Immunotherapy alone offers limited efficacy, but it is still unknown whether its combination with chemotherapy could offer synergistic anti-tumor effects. This phase Ib/II study evaluated the safety and efficacy of combining toripalimab with the gemcitabine plus nab-paclitaxel (GnP) regimen as first-line treatment for locally advanced or metastatic PDAC and explored predictive biomarkers (ChiCTR2000032293). The primary endpoints were safety and overall survival (OS). The secondary outcomes were objective response rate (ORR), disease control rate (DCR), and progression-free survival (PFS). Immune-related biomarkers including programmed death-ligand 1 (PD-L1) expression, genetic status, cytokine levels, and spatial features of the tumor immune microenviroment (TIME) were investigated. Neither serious treatment-related adverse events nor grade 4 immune-related adverse events were reported. Among the 72 patients, the median OS was 8.9 months, 12-month OS rate was 31.9%, with median PFS of 5.6 months, ORR of 33.3%, and DCR of 90.3%. Higher PD-L1 expression, without liver metastases were associated with higher ORR, however these factors could not effectively distinguish responders and non-responders. Importantly, dendritic cells - T helper cells - cytotoxic T lymphocytes (DC-Th-CTL) enriched immune niche and their spatial interactions were dominant predictors of response based on TIME analysis using a cyclic multiplex tissue staining assay, with an area under the curve value of 0.8. Overall, GnP plus toripalimab exhibited good safety and differentiated efficacy in selected population, and the spatial interactions of DC-Th-CTL represent promising predictors to efficacy of immunochemotherapy in locally advanced or metastatic PDAC.
Collapse
Affiliation(s)
- Ke Cheng
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoying Li
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wanrui Lv
- Department of Oncology, Meishan City People's Hospital, Meishan, Sichuan, China
| | - Gang Zhao
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruihan Zhou
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University and Collaborative Innovation Center, West China Hospital, Chengdu, Sichuan, China
| | - Chen Chang
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Heqi Yang
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruizhen Li
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiping Li
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ye Chen
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cheng Yi
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ouying Yan
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chaoxin Xiao
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University and Collaborative Innovation Center, West China Hospital, Chengdu, Sichuan, China
| | - Yi Zhang
- Pancreatic Division, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of General Surgery, ChengDu ShangJing NanFu Hospital, Chengdu, Sichuan, China
| | - Junjie Xiong
- Pancreatic Division, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zixin Huang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Radiology, West China Tianfu Hospital, Sichuan University, Chengdu, Sichuan, China
| | | | - Xin You
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenhao Guo
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Du He
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenwu Ling
- Department of Medical Ultrasound, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Rui Wang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bole Tian
- Pancreatic Division, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Chengjian Zhao
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University and Collaborative Innovation Center, West China Hospital, Chengdu, Sichuan, China.
| | - Dan Cao
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
127
|
Martin RCG, White RR, Bilimoria MM, Kluger MD, Iannitti DA, Polanco PM, Hammil CW, Cleary SP, Heithaus RE, Welling T, Chan CHF. Effectiveness and Safety of Irreversible Electroporation When Used for the Ablation of Stage 3 Pancreatic Adenocarcinoma: Initial Results from the DIRECT Registry Study. Cancers (Basel) 2024; 16:3894. [PMID: 39682087 DOI: 10.3390/cancers16233894] [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: 10/14/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Overall survival for patients with Stage 3 pancreatic ductal adenocarcinoma (PDAC) remains limited, with a median survival of 12 to 15 months. Irreversible electroporation (IRE) is a local tumor ablation method that induces cancerous cell death by disrupting cell membrane homeostasis. The DIRECT Registry study was designed to assess the effectiveness and safety of IRE when combined with standard of care (SOC) treatment for Stage 3 PDAC versus SOC alone in a real-world setting after at least 3 months of induction chemotherapy; Methods: Patients with Stage 3 PDAC treated with IRE plus SOC or SOC alone were prospectively enrolled in a multicenter registry study. Enrollment required 3 months of active multi-agent chemotherapy with no progression before enrollment. Endpoints were 30- and 90-day mortality and adverse events (AEs). RESULTS Eighty-seven IRE and 27 SOC subjects were enrolled in the registry. Mean ages were 64.0 ± 8.4 and 66.4 ± 9.9 years, and mean anterior/posterior tumor diameters were 2.2 ± 0.7 cm and 3.2 ± 1.3 for the IRE and SOC groups respectively (p = 0.0066). All IRE procedures were performed using an open approach. The 90-day all-cause mortality was 5/83 (6.0%) and 2/27 (7.4%) for the IRE and SOC groups, respectively. Two subjects in the IRE group died from treatment-related complications, and one patient in the SOC group died due to chemotherapy-related complications. CONCLUSIONS Initial results from the DIRECT registry study indicate the use of IRE for curative intent tumor ablation in combination with induction chemotherapy has equivalent morbidity and mortality rates when compared to standard-of-care chemotherapy alone.
Collapse
Affiliation(s)
- Robert C G Martin
- Division of Surgical Oncology, Department of Surgery, University of Louisville, Louisville, KY 40202, USA
| | - Rebekah Ruth White
- San Diego Moores Cancer Center, University of California, La Jolla, CA 92037, USA
| | | | - Michael D Kluger
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
| | - David A Iannitti
- Atrium Health Carolinas Medical Center, Charlotte, NC 28203, USA
| | | | - Chet W Hammil
- Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Sean P Cleary
- Division of Surgical Oncology, Department of Surgery, University of Louisville, Louisville, KY 40202, USA
- Mayo Clinic, Rochester, MN 55905, USA
- University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Robert Evans Heithaus
- Department of Radiology, College of Medicine, University of South Florida, Tampa, FL 33606, USA
| | - Theodore Welling
- Division of Surgical Oncology, Department of Surgery, University of Louisville, Louisville, KY 40202, USA
- NYU Langone Health, New York, NY 10016, USA
- UC San Diego Health, San Diego, CA 92037, USA
| | - Carlos H F Chan
- University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| |
Collapse
|
128
|
Kohara Y, Yasuda S, Nagai M, Nakamura K, Matsuo Y, Terai T, Doi S, Sakata T, Sho M. Critical Role of Preoperative Hyperglycemia in Association with GFAT-1 Expression in Resected Pancreatic Cancer. Ann Surg Oncol 2024; 31:7713-7721. [PMID: 39107610 DOI: 10.1245/s10434-024-15985-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/23/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Hyperglycemia is involved in malignant transformation of pancreatic cancer via the hexosamine biosynthetic pathway (HBP). However, few studies have verified this mechanism based on clinical data. This study investigated the complementary effects of hyperglycemia and HBP on pancreatic cancer prognosis using detailed clinical data. METHODS The study analyzed data of 477 patients with pancreatic cancer who underwent pancreatectomy between 2006 and 2020. The patients were divided into normoglycemia and hyperglycemia groups based on their HbA1c levels. Immunostaining for glutamine fructose-6-phosphate transaminase-1 (GFAT-1), the rate-limiting enzyme in HBP, CD4, CD8, and Foxp3, was performed to evaluate the association between survival outcomes, HBP, and local tumor immunity. RESULTS Overall survival (OS) was significantly poorer in the hyperglycemia group than in the normoglycemia group (mean survival time [MST]: 35.0 vs. 47.9 months; p = 0.007). The patients in the hyperglycemia group with high GFAT-1 expression had significantly poorer OS than those with low GFAT-1 expression (MST, 49.0 vs. 27.6 months; p < 0.001). However, the prognosis did not differ significantly between the patients with high and low GFAT-1 expression in the normoglycemia group. In addition, the patients with hyperglycemia and high GFAT-1 expression had fewer CD4+ (p = 0.015) and CD8+ (p = 0.017) T cells and a lower CD8+/Foxp3+ ratio (p = 0.032) than those with hyperglycemia and low GFAT-1 expression. CONCLUSIONS The patients with hyperglycemia and high GFAT-1 expression levels had an extremely poor prognosis. Furthermore, the tumors in these patients were characterized as immunologically cold tumors.
Collapse
Affiliation(s)
| | - Satoshi Yasuda
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Minako Nagai
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Kota Nakamura
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Yasuko Matsuo
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Taichi Terai
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Shunsuke Doi
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Takeshi Sakata
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Masayuki Sho
- Department of Surgery, Nara Medical University, Nara, Japan.
| |
Collapse
|
129
|
Yu SY, Shu YP, Bai XH, Yu J, Lu ZP, Jiang KR, Xu Q. Efficiency evaluation of dual-energy CT to predict the postoperative early recurrence of pancreatic ductal adenocarcinoma. Pancreatology 2024; 24:1123-1132. [PMID: 39327124 DOI: 10.1016/j.pan.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/06/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Abstract
OBJECTIVES To evaluate the efficacy of quantitative parameters from dual-energy CT (DECT) and basic CT features in predicting the postoperative early recurrence (ER) of pancreatic ductal adenocarcinoma (PDAC). METHODS In this study, patients with PDAC who underwent radical resection and DECT from 2018 to 2022 were enrolled and categorised into ER and non-ER groups. The clinical data, basic CT features and DECT parameters of all patients were analyzed. Independent predictors of ER were identified with Logistic regression analyses. Three models (model A: basic CT features; model B: DECT parameters; model C: basic CT features + DECT parameters) were established. Receiver operating characteristic curve analysis was utilized to evaluate predictive performance. RESULTS A total of 150 patients were enrolled (ER group: n = 63; non-ER group: n = 87). Rim enhancement (odds ratio [OR], 3.32), peripancreatic strands appearance (OR, 2.68), electron density in the pancreatic parenchymal phase (P-Rho; OR, 0.90), arterial enhancement fraction (AEF; OR, 0.05) and pancreatic parenchyma fat fraction in the delayed phase (OR, 1.25) were identified as independent predictors of ER. Model C showed the highest area under the curve of 0.898. In addition, the corresponding ER risk factors were identified separately for resectable and borderline resectable PDAC subgroups. CONCLUSIONS DECT quantitative parameters allow for the noninvasive prediction of postoperative ER in patients with PDAC, and the combination of DECT parameters and basic CT features shows a high prediction efficiency. Our model can help to identify patients with high-risk factors to guide preoperative decision making.
Collapse
Affiliation(s)
- Si-Yao Yu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, No 300, Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Yu-Ping Shu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, No 300, Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Xiao-Han Bai
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, No 300, Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Jing Yu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, No 300, Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Zi-Peng Lu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, No 300, Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Kui-Rong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, No 300, Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Qing Xu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, No 300, Guangzhou Road, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
130
|
Liu J, Sidiqi B, McComas K, Gogineni E, Andraos T, Crane CH, Chang DT, Goodman KA, Hall WA, Hoffe S, Mahadevan A, Narang AK, Lee P, Williams TM, Chuong MD. SBRT for Pancreatic Cancer: A Radiosurgery Society Case-Based Practical Guidelines to Challenging Cases. Pract Radiat Oncol 2024; 14:555-573. [PMID: 38986901 DOI: 10.1016/j.prro.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/12/2024]
Abstract
The use of radiation therapy (RT) for pancreatic cancer continues to be controversial, despite recent technical advances. Improvements in systemic control have created an evolving role for RT and the need for improved local tumor control, but currently, no standardized approach exists. Advances in stereotactic body RT, motion management, real-time image guidance, and adaptive therapy have renewed hopes of improved outcomes in this devastating disease with one of the lowest survival rates. This case-based guide provides a practical framework for delivering stereotactic body RT for locally advanced pancreatic cancer. In conjunction with multidisciplinary care, an intradisciplinary approach should guide treatment of the high-risk cases outlined within these guidelines for prospective peer review and treatment safety discussions.
Collapse
Affiliation(s)
- Jason Liu
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California.
| | - Baho Sidiqi
- Department of Radiation Oncology, Northwell Health Cancer Institute, New Hyde Park, New York
| | - Kyra McComas
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, Tennesse
| | - Emile Gogineni
- Department of Radiation Oncology, Ohio State James Cancer Center, Columbus, Ohio
| | - Therese Andraos
- Department of Radiation Oncology, Ohio State James Cancer Center, Columbus, Ohio
| | - Christopher H Crane
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York City, New York
| | - Daniel T Chang
- Department of Radiation Oncology, University of Michigan Health, Ann Arbor, Michigan
| | - Karyn A Goodman
- Department of Radiation Oncology, Mount Sinai Health, New York City, New York
| | - William A Hall
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sarah Hoffe
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Anand Mahadevan
- Department of Radiation Oncology, NYU Langone Health, New York City, New York
| | - Amol K Narang
- Department of Radiation Oncology, Johns Hopkins University Kimmel Cancer Center, Baltimore, Maryland
| | - Percy Lee
- Department of Radiation Oncology, City of Hope Lennar Cancer Center, Irvine, California
| | - Terence M Williams
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California
| | - Michael D Chuong
- Department of Radiation Oncology, Baptist Health South Florida, Miami, Florida
| |
Collapse
|
131
|
Stoop TF, Augustinus S, Björnsson B, Tingstedt B, Andersson B, Wolfgang CL, Werner J, Johansen K, Stommel MWJ, Katz MHG, Ghadimi M, House MG, Ghorbani P, Molenaar IQ, de Wilde RF, Mieog JSD, Keck T, Wellner UF, Uhl W, Besselink MG, Pitt HA, Del Chiaro M. Surgical Outcome After Distal Pancreatectomy With and Without Portomesenteric Venous Resection in Patients with Pancreatic Adenocarcinoma: A Transatlantic Evaluation of Patients in North America, Germany, Sweden, and The Netherlands (GAPASURG). Ann Surg Oncol 2024; 31:8327-8339. [PMID: 39120839 PMCID: PMC11467095 DOI: 10.1245/s10434-024-15932-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Pancreatic adenocarcinoma located in the pancreatic body might require a portomesenteric venous resection (PVR), but data regarding surgical risks after distal pancreatectomy (DP) with PVR are sparse. Insight into additional surgical risks of DP-PVR could support preoperative counseling and intraoperative decision making. This study aimed to provide insight into the surgical outcome of DP-PVR, including its potential risk elevation over standard DP. METHODS We conducted a retrospective, multicenter study including all patients with pancreatic adenocarcinoma who underwent DP ± PVR (2018-2020), registered in four audits for pancreatic surgery from North America, Germany, Sweden, and The Netherlands. Patients who underwent concomitant arterial and/or multivisceral resection(s) were excluded. Predictors for in-hospital/30-day major morbidity and mortality were investigated by logistic regression, correcting for each audit. RESULTS Overall, 2924 patients after DP were included, of whom 241 patients (8.2%) underwent DP-PVR. Rates of major morbidity (24% vs. 18%; p = 0.024) and post-pancreatectomy hemorrhage grade B/C (10% vs. 3%; p = 0.041) were higher after DP-PVR compared with standard DP. Mortality after DP-PVR and standard DP did not differ significantly (2% vs. 1%; p = 0.542). Predictors for major morbidity were PVR (odds ratio [OR] 1.500, 95% confidence interval [CI] 1.086-2.071) and conversion from minimally invasive to open surgery (OR 1.420, 95% CI 1.032-1.970). Predictors for mortality were higher age (OR 1.087, 95% CI 1.045-1.132), chronic obstructive pulmonary disease (OR 4.167, 95% CI 1.852-9.374), and conversion from minimally invasive to open surgery (OR 2.919, 95% CI 1.197-7.118), whereas concomitant PVR was not associated with mortality. CONCLUSIONS PVR during DP for pancreatic adenocarcinoma in the pancreatic body is associated with increased morbidity, but can be performed safely in terms of mortality.
Collapse
Affiliation(s)
- Thomas F Stoop
- Department of Surgery, Division of Surgical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Amsterdam, The Netherlands.
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet at Karolinska University Hospital, Stockholm, Sweden.
| | - Simone Augustinus
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Bergthor Björnsson
- Department of Surgery in Linköping and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Bobby Tingstedt
- Department of Clinical Sciences Lund, Surgery, Skåne University Hospital, Lund University, Lund, Sweden
| | - Bodil Andersson
- Department of Clinical Sciences Lund, Surgery, Skåne University Hospital, Lund University, Lund, Sweden
| | - Christopher L Wolfgang
- Division of Surgical Oncology, Department of Surgery, New York University Medical Center, New York City, NY, USA
| | - Jens Werner
- Department of General, Visceral and Transplant Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Karin Johansen
- Department of Surgery in Linköping and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Martijn W J Stommel
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Ghadimi
- Department of General and Visceral Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Michael G House
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Poya Ghorbani
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet at Karolinska University Hospital, Stockholm, Sweden
| | - I Quintus Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht/St. Antonius Ziekenhuis Nieuwegein, Utrecht & Nieuwegein, The Netherlands
| | - Roeland F de Wilde
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Tobias Keck
- DGAV StuDoQ|Pancreas and Clinic of Surgery, UKSH Campus Lübeck, Lübeck, Germany
| | - Ulrich F Wellner
- DGAV StuDoQ|Pancreas and Clinic of Surgery, UKSH Campus Lübeck, Lübeck, Germany
| | - Waldemar Uhl
- Department of Surgery, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Marc G Besselink
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Henry A Pitt
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.
| | - Marco Del Chiaro
- Department of Surgery, Division of Surgical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
132
|
Ross SB, Popover J, Sucandy I, Christodoulou M, Pattilachan TM, Rosemurgy AS. The Oncological Stress Test of Neoadjuvant Therapy: A Systematic Review in Outcomes of Neoadjuvant Therapy Compared to Upfront Resection Approach for Borderline Resectable Pancreatic Adenocarcinoma. Am Surg 2024; 90:3061-3073. [PMID: 38635295 DOI: 10.1177/00031348241248703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Pancreatic adenocarcinoma, increasingly diagnosed in the United States, has a disheartening initial resection rate of 15%. Neoadjuvant therapy, particularly FOLFIRINOX and gemcitabine-based regimens, is gaining favor for its potential to improve resectability rates and achieving microscopically negative margins (R0) in borderline resectable cases, marked by intricate arterial or venous involvement. Despite surgery being the sole curative approach, actual benefit of neoadjuvant therapy remains debatable. This study scrutinizes current literature on oncological outcomes post-resection of borderline resectable pancreatic cancer. A MEDLINE/PubMed search was conducted to systematically compare oncological outcomes of patients treated with either neoadjuvant therapy with intent of curative resection or an "upfront resection" approach. A total of 1293 studies were initially screened and 30 were included (n = 1714) in this analysis. All studies included data on outcomes of patients with borderline resectable pancreatic adenocarcinoma being treated with neoadjuvant therapy (n = 1387) or a resection-first approach (n = 356). Patients treated with neoadjuvant therapy underwent resection 52% of the time, achieving negative margins of 43% (n = 601). Approximately 77% of patients who received an upfront resection underwent a successful resection, with 39% achieving negative margins. Neoadjuvant therapy remains marginally efficacious in treatment of borderline resectable pancreatic adenocarcinoma, as patients undergo an operation and successful resection less often when treated with neoadjuvant therapy. Rates of curative resection are comparable, despite neoadjuvant therapy being a primary recommendation in borderline resectable cases and employed more often than upfront resection. Upfront resection may offer improved resection rates by intention-to-treat, which can provide more patients with paths to curative resection.
Collapse
Affiliation(s)
- Sharona B Ross
- Digestive Health Institute, AdventHealth Tampa, Tampa, FL, USA
| | - Jesse Popover
- Digestive Health Institute, AdventHealth Tampa, Tampa, FL, USA
| | - Iswanto Sucandy
- Digestive Health Institute, AdventHealth Tampa, Tampa, FL, USA
| | | | | | | |
Collapse
|
133
|
McDonald HG, Kennedy A, Solomon AL, Williams CM, Reagan AM, Cassim E, Harper M, Burke E, Armstrong T, Gosky M, Cavnar M, Pandalai PK, Barry-Hundeyin M, Patel R, Nutalapati S, Moss J, Hull PC, Kolesar J, Pickarski JC, Kim J. Development of a Novel Protocol for Germline Testing in Pancreatic Cancer. Ann Surg Oncol 2024; 31:7705-7712. [PMID: 39133448 DOI: 10.1245/s10434-024-16011-3] [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: 05/20/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Guidelines now recommend universal germline genetic testing (GGT) for all pancreatic ductal adenocarcinoma (PDAC) patients. Testing provides information on actionable pathogenic variants and guides management of patients and family. Since traditional genetic counseling (GC) models are time-intensive and GC resources are sparse, new approaches are needed to comply with guidelines without overwhelming available resources. METHODS A novel protocol was developed for physician-led GGT. Completed test kits were delivered to the GC team, who maintained a prospective database and mailed all orders. If results revealed pathogenic variants for PDAC, patients were offered comprehensive GC, whereas negative and variant of uncertain significance (VUS) test results were reported to patients via brief calls. RESULTS During protocol implementation between January 2020 and December 2022, 310 (81.5%) patients underwent GGT, with a physician compliance rate of 82.6% and patient compliance rate of 98.7%. Of 310 patients tested, 44 (14.2%) patients had detection of pathogenic variants, while 83 (26.8%) patients had VUS. Pathogenic variants included BRCA1/BRCA2/PALB2 (n = 18, 5.8%), ATM (n = 9, 2.9%), CFTR (n = 4, 1.3%), EPCAM/MLH1/MSH2/MSH6/PMS2 (n = 3, 1.0%), and CDKN2A (n = 2, 0.7%). The GC team successfully contacted all patients with pathogenic variants to discuss results and offer comprehensive GC. CONCLUSION Our novel protocol facilitated GGT with excellent compliance despite limited GC resources. This framework for GGT allocates GC resources to those patients who would benefit most from GC. As we continue to expand the program, we seek to implement methods to ensure compliance with cascade testing of high-risk family members.
Collapse
Affiliation(s)
- Hannah G McDonald
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Andrew Kennedy
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Angelica L Solomon
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Chelsey M Williams
- Division of Hematology Oncology, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Anna M Reagan
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Emily Cassim
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Megan Harper
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Erin Burke
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Terra Armstrong
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Michael Gosky
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Michael Cavnar
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Prakash K Pandalai
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Mautin Barry-Hundeyin
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Reema Patel
- Division of Hematology Oncology, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Snigdha Nutalapati
- Division of Hematology Oncology, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Jessica Moss
- Division of Hematology Oncology, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Pamela C Hull
- Department of Behavioral Science, University of Kentucky, Lexington, KY, USA
| | - Jill Kolesar
- College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | | | - Joseph Kim
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
134
|
Dong D, Yu X, Xu J, Yu N, Liu Z, Sun Y. Cellular and molecular mechanisms of gastrointestinal cancer liver metastases and drug resistance. Drug Resist Updat 2024; 77:101125. [PMID: 39173439 DOI: 10.1016/j.drup.2024.101125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024]
Abstract
Distant metastases and drug resistance account for poor survival of patients with gastrointestinal (GI) malignancies such as gastric cancer, pancreatic cancer, and colorectal cancer. GI cancers most commonly metastasize to the liver, which provides a unique immunosuppressive tumour microenvironment to support the development of a premetastatic niche for tumor cell colonization and metastatic outgrowth. Metastatic tumors often exhibit greater resistance to drugs than primary tumors, posing extra challenges in treatment. The liver metastases and drug resistance of GI cancers are regulated by complex, intertwined, and tumor-dependent cellular and molecular mechanisms that influence tumor cell behavior (e.g. epithelial-to-mesenchymal transition, or EMT), tumor microenvironment (TME) (e.g. the extracellular matrix, cancer-associated fibroblasts, and tumor-infiltrating immune cells), tumor cell-TME interactions (e.g. through cytokines and exosomes), liver microenvironment (e.g. hepatic stellate cells and macrophages), and the route and mechanism of tumor cell dissemination (e.g. circulating tumor cells). This review provides an overview of recent advances in the research on cellular and molecular mechanisms that regulate liver metastases and drug resistance of GI cancers. We also discuss recent advances in the development of mechanism-based therapy for these GI cancers. Targeting these cellular and molecular mechanisms, either alone or in combination, may potentially provide novel approaches to treat metastatic GI malignancies.
Collapse
Affiliation(s)
- Daosong Dong
- Department of Pain, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Xue Yu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Molecular Pathology and Epidemiology of Gastric Cancer in the Universities of Liaoning Province, Shenyang, Liaoning 110001, China
| | - Jingjing Xu
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Na Yu
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Zhe Liu
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Yanbin Sun
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
135
|
Gupta A, De Jesus-Acosta A, Le D, Pishvaian M, Zaidi N, Zheng L, Laheru D. Clinical outcomes of liposomal irinotecan in patients with advanced pancreatic cancer previously treated with conventional irinotecan: A meta-analysis of real-world evidence. Cancer 2024; 130:3734-3744. [PMID: 38985839 DOI: 10.1002/cncr.35479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Pivotal clinical trials supported survival benefits of liposomal irinotecan (nal-IRI) plus fluorouracil/leucovorin in patients with pancreatic ductal adenocarcinoma (PDAC) who previously received gemcitabine-based therapy. There are concerns about the benefits of nal-IRI in patients who received FOLFIRINOX (combined fluorouracil, leucovorin, IRI, and oxaliplatin) because of potential cross-resistance to IRI. The objective of this meta-analysis was to characterize the impact of the previous receipt of IRI on the outcomes of nal-IRI regimens in patients with advanced PDAC. METHODS Real-world studies evaluating the outcomes of nal-IRI in patients who had prior IRI exposure published up to April 2023 were searched using electronic databases. The meta-analysis was conducted using a random effects model to estimate pooled hazard ratios (HRs) and 95% confidence intervals (CIs). RESULTS Eight studies (n = 1368 patients) were included. The pooled median progression-free survival (PFS) was 2.02 months (95% CI, 1.43-2.57 months), and the median overall survival (OS) was 4.26 months (95% CI, 3.03-5.39 months). Patients with prior IRI exposure had PFS (HR, 1.17; 95% CI, 0.94-1.47; p = .17) and OS (HR, 1.16; 95% CI, 0.95-1.42; p = .16) comparable to patients without prior IRI exposure. Likewise, patients who had progressive disease on conventional IRI had PFS (HR, 1.50; 95% CI, 0.73-3.08; p = .24) and OS (HR, 1.70; 95% CI, 0.68-4.27; p = .26) with nal-IRI comparable to patients who had no progressive disease. CONCLUSIONS Prior IRI exposure does not affect the survival outcomes of nal-IRI regimens in patients who have advanced PDAC. The selection of later lines of chemotherapy regimens should be based on the differential safety profile, patient status, the cost of treatment, and health-related quality of life.
Collapse
Affiliation(s)
- Amol Gupta
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Ana De Jesus-Acosta
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Dung Le
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Michael Pishvaian
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Neeha Zaidi
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Lei Zheng
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Daniel Laheru
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| |
Collapse
|
136
|
Pan Y, Ran T, Zhang X, Qin X, Zhang Y, Zhou C, Zou D. Adequacy of EUS-guided fine-needle aspiration and fine-needle biopsy for next-generation sequencing in pancreatic malignancies: A systematic review and meta-analysis. Endosc Ultrasound 2024; 13:366-375. [PMID: 39802109 PMCID: PMC11723693 DOI: 10.1097/eus.0000000000000097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2025] Open
Abstract
Background and Objectives A majority of pancreatic malignancies are unresectable at the time of presentation and require EUS-guided fine-needle aspiration or fine-needle biopsy (EUS-FNA/FNB) for diagnosis. With the advent of precision therapy, there is an increasing need to use EUS-FNA/FNB sample for genetic analysis. Next-generation sequencing (NGS) is a preferred technology to detect genetic mutations with high sensitivity in small specimens. We performed a meta-analysis to evaluate the adequacy of EUS-FNA/FNB for NGS in pancreatic malignancies. Methods PubMed, Embase, Cochrane Library, and Web of Science were searched from database inception to November 11, 2023. The primary outcome was the proportion of sufficient sample acquired by EUS-FNA/FNB in pancreatic malignancies for NGS. Secondary outcomes were the proportion of sufficient sample for NGS in pancreatic ductal adenocarcinoma (PDAC) and the detection rates of mutations in KRAS, TP53, CDKN2A, and SMAD4 and actionable mutations in PDAC. The pooled proportions were calculated using a random-effects model. Potential sources of heterogeneity were investigated with subgroup analyses and meta-regression. Results Twenty studies with 881 samples were included. The pooled adequacy of EUS-FNA/FNB sample for NGS was 89.9% (95% CI, 80.8%-96.7%) in pancreatic malignancies and 92.0% (95% CI, 81.3%-98.8%) in PDAC. Screening sample suitability before NGS testing was associated with lower adequacy in subgroup analysis (79.7% vs. 98.4%, P = 0.001). The pooled prevalences of mutations in KRAS, TP53, CDKN2A, and SMAD4 in PDAC were 87.4% (95% CI, 83.2%-91.2%), 62.6% (95% CI, 53.2%-71.7%), 20.6% (95% CI, 11.9%-30.8%), and 19.4% (95% CI, 11.2%-29.1%), respectively. The pooled prevalence of potentially actionable mutations in PDAC was 14.5% (95% CI, 8.2%-22.0%). Conclusions In the majority of cases, EUS-FNA/FNB can acquire adequate sample for NGS and identify tumor-specific mutations in patients with pancreatic malignancies. Strict pre-analysis screening criteria may negatively impact the sample adequacy and the success rate for NGS.
Collapse
Affiliation(s)
| | | | | | | | | | - Chunhua Zhou
- Department of Gastroenterology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Duowu Zou
- Department of Gastroenterology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| |
Collapse
|
137
|
Wong P, Pollini T, Adam MA, Alseidi A, Corvera CU, Hirose K, Kirkwood KS, Nakakura EK, Thornblade L, Maker AV. Distinct Indications for Adjuvant Therapy in Resected Invasive Mucinous Cystic Neoplasms of the Pancreas Compared with Pancreatic Ductal Adenocarcinoma. Ann Surg Oncol 2024; 31:8276-8286. [PMID: 39068306 PMCID: PMC11467002 DOI: 10.1245/s10434-024-15841-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 06/24/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Surgical and adjuvant management of mucinous cystic neoplasms (MCNs) lacks formal guidelines and data is limited to institutional studies. Factors associated with receipt of adjuvant therapy and any associated impact on survival remain to be clarified. In the absence of other data, guidelines that recommend adjuvant chemotherapy for invasive pancreatic adenocarcinoma have been extrapolated to MCN. PATIENTS AND METHODS The National Cancer Database (2004-2019) was utilized to identify all patients that underwent pancreatic resection for invasive MCNs. Patients that received neoadjuvant therapy or did not undergo lymphadenectomy were excluded. Patient, tumor, and treatment factors associated with survival were assessed. RESULTS For 161 patients with invasive MCN, median overall survival (OS) was 133 months and 45% of patients received adjuvant therapy. Multivariable analysis demonstrated that poorly differentiated tumors [odds ratio (OR) 4.19, 95% confidence interval (CI) 1.47-11.98; p = 0.008] and positive lymph node status (OR 2.67, 95% CI 1.02-6.98; p = 0.042) were independent predictors of receiving adjuvant therapy. Lymph node positivity [hazard ratio (HR) 2.90, 95% CI 1.47-5.73; p = 0.002], positive margins (HR 5.28, 95% CI 2.28-12.27; p < 0.001), and stage III disease (HR 12.46, 95% CI 1.40-111.05; p = 0.024) were associated with worse OS. Receipt of adjuvant systemic therapy was independently associated with decreased risk of mortality in node positive patients (HR 0.23, 95% CI 0.10-0.69; p = 0.002). Survival was not associated with adjuvant therapy in patients with negative lymph nodes or margin negative status. CONCLUSION In contrast to pancreatic ductal adenocarcinoma (PDAC), where adjuvant therapy improves OS for every tumor stage, surgery alone for invasive MCN is not associated with improved OS compared with surgery plus adjuvant therapy in node-negative patients. Surgery alone is likely sufficient for a subset of invasive MCN.
Collapse
Affiliation(s)
- Paul Wong
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, USA
| | - Tommaso Pollini
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, USA
| | - Mohamed A Adam
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, USA
| | - Adnan Alseidi
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, USA
| | - Carlos U Corvera
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, USA
| | - Kenzo Hirose
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, USA
| | - Kimberly S Kirkwood
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, USA
| | - Eric K Nakakura
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, USA
| | - Lucas Thornblade
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, USA
| | - Ajay V Maker
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, USA.
| |
Collapse
|
138
|
Marzioni M, Crinò SF, Lisotti A, Fuccio L, Vanella G, Amato A, Bertani H, Binda C, Coluccio C, Forti E, Fugazza A, Ligresti D, Maida M, Marchegiani G, Mauro A, Mirante VG, Ricci C, Rizzo GEM, Scimeca D, Spadaccini M, Arvanitakis M, Anderloni A, Fabbri C, Tarantino I, Arcidiacono PG. Biliary drainage in patients with malignant distal biliary obstruction: results of an Italian consensus conference. Surg Endosc 2024; 38:6207-6226. [PMID: 39317905 PMCID: PMC11525304 DOI: 10.1007/s00464-024-11245-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND Malignant Distal Biliary Obstruction (MBDO) is a common event occurring along the natural history of both pancreatic cancer and cholangiocarcinoma. Epidemiological and biological features make MBDO one of the key elements of the clinical management of patients suffering for of pancreatic cancer or cholangiocarcinoma. The development of dedicated biliary lumen-apposing metal stents (LAMS) is changing the clinical work up of patients with MBDO. i-EUS is an Italian network of clinicians and scientists with a special interest in biliopancreatic endoscopy, EUS in particular. METHODS The scientific methodology was chosen in line with international guidance and in a fashion similar to those applied by broader scientific associations. PICO questions were elaborated and subsequently voted by a broad panel of experts within a simplified Delphi process. RESULTS AND CONCLUSIONS The manuscripts describes the results of a consensus conference organized by i-EUS with the aim of providing an evidence based-guidance for the appropriate use of the techniques in patients with MBDO.
Collapse
Affiliation(s)
- Marco Marzioni
- Clinic of Gastroenterology and Hepatology, Università Politecnica delle Marche - Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy.
| | - Stefano Francesco Crinò
- Diagnostic and Interventional Endoscopy of Pancreas, The Pancreas Institute, G.B. Rossi University Hospital, 37134, Verona, Italy
| | - Andrea Lisotti
- Gastroenterology Unit, Hospital of Imola, University of Bologna, Imola, Italy
| | - Lorenzo Fuccio
- Department of Medical and Surgical Sciences, University of Bologna - Gastroenterology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, S. Orsola Hospital, Bologna, Italy
| | - Giuseppe Vanella
- Pancreatobiliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Institute, Milan, Italy
| | - Arnaldo Amato
- Department of Digestive Endoscopy and Gastroenterology ASST, Lecco, Italy
| | - Helga Bertani
- Gastroenterologia ed Endoscopia Digestiva Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Cecilia Binda
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, AUSL Romagna, Forlì-Cesena, Italy
| | - Chiara Coluccio
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, AUSL Romagna, Forlì-Cesena, Italy
| | - Edoardo Forti
- Digestive and Interventional Endoscopy Unit, ASST Niguarda Hospital, Milan, Italy
| | - Alessandro Fugazza
- Division of Gastroenterology and Digestive Endoscopy, Humanitas Research Hospital - IRCCS, Rozzano, 20089, Milan, Italy
| | - Dario Ligresti
- Endoscopy Service, Department of Diagnostic and Therapeutic Services, IRCCS - ISMETT, Palermo, Italy
| | - Marcello Maida
- Gastroenterology Unit, Umberto I Hospital - Department of Medicine and Surgery, University of Enna 'Kore', Enna, Italy
| | - Giovanni Marchegiani
- Department of Surgical Oncological and Gastroenterological Sciences, Padua University Hospital, Padua, Italy
| | - Aurelio Mauro
- Gastroenterology and Digestive Endoscopy Unit, IRCCS Foundation Policlinico San Matteo, Viale Camillo Golgi 19, 27100, Pavia, Italy
| | - Vincenzo Giorgio Mirante
- Gastroenterologia ed Endoscopia Digestiva, Dipartimento Oncologico e Tecnologie Avanzate, AUSL IRCCS Reggio Emilia, Reggio Emilia, Italy
| | - Claudio Ricci
- Department of Medical and Surgical Sciences, University of Bologna - Division of Pancreatic Surgery, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | | | - Daniela Scimeca
- Gastroenterology and Endoscopy Unit, ARNAS Civico - Di Cristina - Benfratelli Hospital, 90127, Palermo, Italy
| | - Marco Spadaccini
- Division of Gastroenterology and Digestive Endoscopy, Humanitas Research Hospital - IRCCS, Rozzano, 20089, Milan, Italy
| | - Marianna Arvanitakis
- Clinic of Gastroenterology and Hepatology, Università Politecnica delle Marche - Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy
- Diagnostic and Interventional Endoscopy of Pancreas, The Pancreas Institute, G.B. Rossi University Hospital, 37134, Verona, Italy
- Gastroenterology Unit, Hospital of Imola, University of Bologna, Imola, Italy
- Department of Medical and Surgical Sciences, University of Bologna - Gastroenterology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, S. Orsola Hospital, Bologna, Italy
- Pancreatobiliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Institute, Milan, Italy
- Department of Digestive Endoscopy and Gastroenterology ASST, Lecco, Italy
- Gastroenterologia ed Endoscopia Digestiva Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, AUSL Romagna, Forlì-Cesena, Italy
- Digestive and Interventional Endoscopy Unit, ASST Niguarda Hospital, Milan, Italy
- Division of Gastroenterology and Digestive Endoscopy, Humanitas Research Hospital - IRCCS, Rozzano, 20089, Milan, Italy
- Endoscopy Service, Department of Diagnostic and Therapeutic Services, IRCCS - ISMETT, Palermo, Italy
- Gastroenterology Unit, Umberto I Hospital - Department of Medicine and Surgery, University of Enna 'Kore', Enna, Italy
- Department of Surgical Oncological and Gastroenterological Sciences, Padua University Hospital, Padua, Italy
- Gastroenterology and Digestive Endoscopy Unit, IRCCS Foundation Policlinico San Matteo, Viale Camillo Golgi 19, 27100, Pavia, Italy
- Gastroenterologia ed Endoscopia Digestiva, Dipartimento Oncologico e Tecnologie Avanzate, AUSL IRCCS Reggio Emilia, Reggio Emilia, Italy
- Department of Medical and Surgical Sciences, University of Bologna - Division of Pancreatic Surgery, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
- Gastroenterology and Endoscopy Unit, ARNAS Civico - Di Cristina - Benfratelli Hospital, 90127, Palermo, Italy
| | - Andrea Anderloni
- Gastroenterology and Digestive Endoscopy Unit, IRCCS Foundation Policlinico San Matteo, Viale Camillo Golgi 19, 27100, Pavia, Italy
| | - Carlo Fabbri
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, AUSL Romagna, Forlì-Cesena, Italy
| | - Ilaria Tarantino
- Endoscopy Service, Department of Diagnostic and Therapeutic Services, IRCCS - ISMETT, Palermo, Italy
| | - Paolo Giorgio Arcidiacono
- Pancreatobiliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Institute, Milan, Italy
| |
Collapse
|
139
|
Archasappawat S, Al-Musawi F, Liu P, Lee E, Hwang CI. Familial Pancreatic Cancer Research: Bridging Gaps in Basic Research and Clinical Application. Biomolecules 2024; 14:1381. [PMID: 39595558 PMCID: PMC11592027 DOI: 10.3390/biom14111381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/07/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Familial pancreatic cancer (FPC) represents a significant yet underexplored area in pancreatic cancer research. Basic research efforts are notably limited, and when present, they are predominantly centered on the BRCA1 and BRCA2 mutations due to the scarcity of other genetic variants associated with FPC, leading to a limited understanding of the broader genetic landscape of FPC. This review examines the current state of FPC research, focusing on the molecular mechanisms driving pancreatic ductal adenocarcinoma (PDAC) progression. It highlights the role of homologous recombination (HR) and its therapeutic exploitation via synthetic lethality with PARP inhibitors in BRCA1/2-deficient tumors. The review discusses various pre-clinical models of FPC, including conventional two-dimensional (2D) cell lines, patient-derived organoids (PDOs), patient-derived xenografts (PDXs), and genetically engineered mouse models (GEMMs), as well as new advancements in FPC research.
Collapse
Affiliation(s)
- Suyakarn Archasappawat
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
- University of California Davis Comprehensive Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| | - Fatimah Al-Musawi
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
| | - Peiyi Liu
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
| | - EunJung Lee
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
| | - Chang-il Hwang
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
- University of California Davis Comprehensive Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| |
Collapse
|
140
|
Navez J, Pezzullo M, Bouchart C, Arsenijevic T, Demetter P, Closset J, Azurmendi Senar O, Racu ML, D’Haene N, Devière J, Verset L, Bali MA, van Laethem JL. Impact of the radiological morphology of the mesopancreas on the outcome after pancreatoduodenectomy for pancreatic ductal adenocarcinoma: retrospective study. BJS Open 2024; 8:zrae134. [PMID: 39601263 PMCID: PMC11599710 DOI: 10.1093/bjsopen/zrae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/18/2024] [Accepted: 10/09/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The most frequently invaded margins on pancreatoduodenectomy specimens for pancreatic ductal adenocarcinoma are vascular margins, particularly the superior mesenteric artery (or mesopancreatic) margin. Due to limited exploration of the radiological aspect of the mesopancreas, the aim of this study was to evaluate mesopancreatic infiltration through imaging of patients with pancreatic ductal adenocarcinoma who underwent pancreatoduodenectomy, to correlate these findings with histopathology and evaluate their impact on survival. METHODS Data for all patients who underwent pancreatoduodenectomy for pancreatic ductal adenocarcinoma from 2015 to 2021 were reviewed, including review of surgical margin histopathology and blinded review of preoperative diagnostic imaging. According to qualitative radiological assessment, the mesopancreas was characterized as having normal fat, fat stranding, or solid infiltration. Survival data were analysed using Cox regression. RESULTS A total of 149 patients were included. At baseline imaging, mesopancreatic fat stranding or solid infiltration was present in 47 patients (31.5%) and 20 patients (13.4%) respectively. Median overall survival and disease-free survival were significantly lower with mesopancreatic solid infiltration (17 and 8 months) compared with normal fat (30 and 14 months) and fat stranding (29 and 16 months) (P = 0.017 and 0.028 respectively). In multivariable analysis, pathological tumour size was an independent prognostic factor for overall survival, and tumour location in the uncinate process and pathological tumour size were independent prognostic factors for disease-free survival. CONCLUSION At diagnostic imaging, solid infiltration (but not fat stranding) of the mesopancreas is associated with a poor prognosis for pancreatic ductal adenocarcinoma patients who undergo pancreatoduodenectomy. Pathological tumour size significantly influences the prediction of overall survival, and tumour location in the uncinate process and pathological tumour size significantly influence the prediction of disease-free survival, suggesting further exploration of underlying mechanisms related to retroperitoneal tumoral invasion of vascular margins and the mesopancreas.
Collapse
Affiliation(s)
- Julie Navez
- Medico-Surgical Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - Martina Pezzullo
- Department of Radiology, Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - Christelle Bouchart
- Department of Radiotherapy, Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - Tatjana Arsenijevic
- Medico-Surgical Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Pieter Demetter
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Centre de Morphologie Pathologique (CMP), Cerba Path, Brussels, Belgium
| | - Jean Closset
- Medico-Surgical Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - Oier Azurmendi Senar
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Marie-Lucie Racu
- Department of Pathology, Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - Nicky D’Haene
- Department of Pathology, Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - Jacques Devière
- Medico-Surgical Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - Laurine Verset
- Department of Pathology, Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - Maria A Bali
- Department of Radiology, Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - Jean-Luc van Laethem
- Medico-Surgical Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| |
Collapse
|
141
|
He H, Zou CF, Jiang YJ, Yang F, Di Y, Li J, Jin C, Fu DL. Recurrence scoring system predicting early recurrence for patients with pancreatic ductal adenocarcinoma undergoing pancreatectomy and portomesenteric vein resection. World J Gastrointest Surg 2024; 16:3185-3201. [PMID: 39575290 PMCID: PMC11577395 DOI: 10.4240/wjgs.v16.i10.3185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/19/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND Pancreatectomy with concomitant portomesenteric vein resection (PVR) enables patients with portomesenteric vein (PV) involvement to achieve radical resection of pancreatic ductal adenocarcinoma, however, early recurrence (ER) is frequently observed. AIM To predict ER and identify patients at high risk of ER for individualized therapy. METHODS Totally 238 patients undergoing pancreatectomy and PVR were retrospectively enrolled and were allocated to the training or validating cohort. Univariate Cox and LASSO regression analyses were performed to construct serum recurrence score (SRS) based on 26 serum-derived parameters. Uni- and multivariate Cox regression analyses of SRS and 18 clinicopathological variables were performed to establish a Nomogram. Receiver operating characteristic curve analysis was used to evaluate the predictive accuracy. Survival analysis was performed using Kaplan-Meier method and log-rank test. RESULTS Independent serum-derived recurrence-relevant factors of LASSO regression model, including postoperative carbohydrate antigen 19-9, postoperative carcinoembryonic antigen, postoperative carbohydrate antigen 125, preoperative albumin (ALB), preoperative platelet to ALB ratio, and postoperative platelets to lymphocytes ratio, were used to construct SRS [area under the curve (AUC): 0.855, 95%CI: 0.786-0.924]. Independent risk factors of recurrence, including SRS [hazard ratio (HR): 1.688, 95%CI: 1.075-2.652], pain (HR: 1.653, 95%CI: 1.052-2.598), perineural invasion (HR: 2.070, 95%CI: 0.827-5.182), and PV invasion (HR: 1.603, 95%CI: 1.063-2.417), were used to establish the recurrence nomogram (AUC: 0.869, 95%CI: 0.803-0.934). Patients with either SRS > 0.53 or recurrence nomogram score > 4.23 were considered at high risk for ER, and had poor long-term outcomes. CONCLUSION The recurrence scoring system unique for pancreatectomy and PVR, will help clinicians in predicting recurrence efficiently and identifying patients at high risk of ER for individualized therapy.
Collapse
Affiliation(s)
- Hang He
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Cai-Feng Zou
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Yong-Jian Jiang
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Feng Yang
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Yang Di
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Ji Li
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Chen Jin
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - De-Liang Fu
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| |
Collapse
|
142
|
Hong W, Zhang L, Yu Z, Wang Y, Qi Y. Intestinal obstruction following antituberculosis therapy in a patient with pancreatic carcinoma and pulmonary tuberculosis: a case report. J Med Case Rep 2024; 18:495. [PMID: 39434174 PMCID: PMC11494838 DOI: 10.1186/s13256-024-04849-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/10/2024] [Indexed: 10/23/2024] Open
Abstract
INTRODUCTION Intestinal obstruction is a common complication in patients with advanced malignancies, often attributed to the disease itself or as a side effect of opioid analgesics used for pain management. However, the occurrence of intestinal obstruction following antituberculosis therapy is rare. CASE PRESENTATION We report a unique case of a 58-year-old Asian male diagnosed with stage IV pancreatic carcinoma and pulmonary tuberculosis. The patient was initiated on a regimen of ethambutol hydrochloride, pyrazinamide, rifampicin, and isoniazid tablets (II) for tuberculosis, alongside morphine for the management of severe cancer-related pain. Subsequently, he developed symptoms indicative of intestinal obstruction. Despite discontinuation of morphine, the patient's symptoms persisted until he autonomously ceased all medications, leading to a rapid improvement in his condition. This unexpected resolution highlighted the antituberculosis drugs as the probable cause of his intestinal obstruction. CONCLUSIONS This case underscores the importance of considering antituberculosis drugs as a potential cause of intestinal obstruction, especially in patients who do not respond to conventional management strategies for drug-induced gastrointestinal side effects. It also emphasizes the need for heightened vigilance and monitoring when prescribing these medications to patients with advanced malignancies, to promptly identify and address rare but significant side effects.
Collapse
Affiliation(s)
- Wei Hong
- Second People's Hospital of Liaocheng, 306 Health Street, Linqing, Shandong, China
| | - Lei Zhang
- Second People's Hospital of Liaocheng, 306 Health Street, Linqing, Shandong, China
| | - Zunshun Yu
- Second People's Hospital of Liaocheng, 306 Health Street, Linqing, Shandong, China
| | - Yanjun Wang
- Second People's Hospital of Liaocheng, 306 Health Street, Linqing, Shandong, China
| | - Youkun Qi
- Second People's Hospital of Liaocheng, 306 Health Street, Linqing, Shandong, China.
| |
Collapse
|
143
|
Bravo AC, Morão B, Luz A, Dourado R, Oliveira B, Guedes A, Moreira-Barbosa C, Fidalgo C, Mascarenhas-Lemos L, Costa-Santos MP, Maio R, Paulino J, Viana Baptista P, Fernandes AR, Cravo M. Bringing Hope to Improve Treatment in Pancreatic Ductal Adenocarcinoma-A New Tool for Molecular Profiling of KRAS Mutations in Tumor and Plasma Samples. Cancers (Basel) 2024; 16:3544. [PMID: 39456638 PMCID: PMC11506488 DOI: 10.3390/cancers16203544] [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: 09/17/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) incidence is rising, and prognosis remains poor due to late diagnosis and limited effective therapies. Currently, patients are treated based on TNM staging, without molecular tumor characterization. This study aimed to validate a technique that combines the amplification refractory mutation system (ARMS) with high-resolution melting analysis (HRMA) for detecting mutations in codon 12 of KRAS in tumor and plasma, and to assess its prognostic value. METHODS Prospective study including patients with newly diagnosed PDAC with tumor and plasma samples collected before treatment. Mutations in codon 12 of KRAS (G12D, G12V, G12C, and G12R) were detected using ARMS-HRMA and compared to Sanger sequencing (SS). Univariate and multivariate analyses were used to evaluate the prognostic significance of these mutations. RESULTS A total of 88 patients, 93% with ECOG-PS 0-1, 57% with resectable disease. ARMS-HRMA technique showed a higher sensitivity than SS, both in tumor and plasma (77% vs. 51%; 25 vs. 0%, respectively). The most frequent mutation was G12D (n = 32, 36%), followed by G12V (n = 22, 25%). On multivariate analysis, patients with G12D and/or G12C mutations, either in tumor or plasma, had lower PFS (HR 1.792, 95% CI 1.061-3.028, p = 0.029; HR 2.081, 95% CI 1.014-4.272, p = 0.046, respectively) and lower OS (HR 1.757, 95% CI 1.013-3.049, p = 0.045; HR 2.229, 95% CI 1.082-4.594, p = 0.030, respectively). CONCLUSIONS ARMS-HRMA is a rapid and cost-effective method for detecting KRAS mutations in PDAC patients, offering the potential for stratifying prognosis and guiding treatment decisions. The presence of G12D and G12C mutations in both tumor and plasma is associated with a poorer prognosis.
Collapse
Affiliation(s)
- Ana Catarina Bravo
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
| | - Bárbara Morão
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
| | - André Luz
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Rúben Dourado
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Beatriz Oliveira
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Ana Guedes
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
- Hospital da Luz Learning Health, Luz Saúde, 1500-650 Lisboa, Portugal
| | - Catarina Moreira-Barbosa
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
- Hospital da Luz Learning Health, Luz Saúde, 1500-650 Lisboa, Portugal
| | - Catarina Fidalgo
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
| | - Luís Mascarenhas-Lemos
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
- NOVA Medical School, 1169-056 Lisboa, Portugal
- Catolica Medical School, 1649-023 Lisboa, Portugal
| | | | - Rui Maio
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
- NOVA Medical School, 1169-056 Lisboa, Portugal
| | - Jorge Paulino
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
- NOVA Medical School, 1169-056 Lisboa, Portugal
| | - Pedro Viana Baptista
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Alexandra R. Fernandes
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Marília Cravo
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
- Lisbon School of Medicine, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| |
Collapse
|
144
|
Araki T, Kawahira M, Shimokawa M, Otsuka T, Hayashi K, Sonoda Y, Honda T, Nakao K, Shibuki T, Nakazawa J, Arima S, Fukahori M, Miwa K, Koga F, Ueda Y, Kubotsu Y, Makiyama A, Shimokawa H, Takeshita S, Nishikawa K, Komori A, Otsu S, Hosokawa A, Sakai T, Oda H, Arita S, Taguchi H, Tsuneyoshi K, Kawaguchi Y, Fujita T, Sakae T, Nio K, Ide Y, Ureshino N, Shirakawa T, Mizuta T, Mitsugi K. Real-World Analysis of the Correlation between Overall Survival and Progression-Free Survival in Advanced Pancreatic Cancer: Results of NAPOLEON-1 and 2 Studies. Oncology 2024:1-11. [PMID: 39427640 DOI: 10.1159/000542137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/11/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) improve overall survival (OS) and progression-free survival (PFS) in patients with pancreatic cancer, compared with gemcitabine (GEM). However, whether PFS is a surrogate marker of OS in pancreatic cancer chemotherapy focusing on FOLFIRINOX or GEM plus nab-paclitaxel remains unknown. We aimed to verify whether PFS can be a surrogate marker of OS in prognosis prediction. METHODS This was an integrated analysis of the NAPOLEON study and retrospective cohort of the NAPOLEON-2 study - a multicenter observational study conducted in Japan, using real-world data. The primary and secondary endpoints were OS and PFS, respectively. The correlation between OS and PFS in first- and second-line treatments was assessed using Method of Moments estimation. An analysis was performed in patients with confirmed OS at the end of follow-up. The NAPOLEON-2 cohort included only patients who received 5-fluorouracil, leucovorin, and nanoliposomal irinotecan (NFF) as second-line treatment. RESULTS Among 479 patients, the correlation between PFS and OS from first- and second-line chemotherapies was calculated in 310 and 225 patients, respectively. The R-squared values for the correlation between PFS and OS from first- and second-line chemotherapies were 0.74 and 0.76, respectively. There was no statistically significant difference in first-line treatment between the FOLFIRINOX and GEM plus nab-paclitaxel groups (p = 0.92). Therefore, the FOLFIRINOX group may not have shown a stronger correlation than the NFF group. CONCLUSION PFS can be a surrogate marker of OS in first- and second-line therapies. Appropriate prognostic estimation might contribute to proper treatment selection.
Collapse
Affiliation(s)
- Tomonori Araki
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan,
| | - Machiko Kawahira
- Department of Gastroenterology, Kagoshima Kouseiren Hospital, Kagoshima, Japan
| | - Mototsugu Shimokawa
- Clinical Research Institute, National Kyushu Cancer Center, Fukuoka, Japan
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Taiga Otsuka
- Department of Medical Oncology, Saga-Ken Medical Center Koseikan, Saga, Japan
- Department of Internal Medicine, Minato Medical Clinic, Fukuoka, Japan
| | - Kohei Hayashi
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yuki Sonoda
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Taro Shibuki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Junichi Nakazawa
- Department of Medical Oncology, Kagoshima City Hospital, Kagoshima, Japan
| | - Shiho Arima
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Masaru Fukahori
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Keisuke Miwa
- Multidisciplinary Treatment Cancer Center, Kurume University Hospital, Fukuoka, Japan
| | - Futa Koga
- Department of Hepatobiliary and Pancreatology, Saga Medical Center Koseikan, Saga, Japan
| | - Yujiro Ueda
- Department of Hematology and Oncology, Japanese Red Cross Kumamoto Hospital, Kumamoto, Japan
| | - Yoshihito Kubotsu
- Department of Internal Medicine, Karatsu Red Cross Hospital, Saga, Japan
| | - Akitaka Makiyama
- Department of Hematology Oncology, Japan Community Healthcare Organization Kyushu Hospital, Fukuoka, Japan
| | - Hozumi Shimokawa
- Department of Hematology Oncology, Japan Community Healthcare Organization Kyushu Hospital, Fukuoka, Japan
| | - Shigeyuki Takeshita
- Department of Gastroenterology, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Kazuo Nishikawa
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Oita, Japan
| | - Azusa Komori
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Oita, Japan
| | - Satoshi Otsu
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Oita, Japan
| | - Ayumu Hosokawa
- Department of Clinical Oncology, University of Miyazaki Hospital, Miyazaki, Japan
| | - Tatsunori Sakai
- Department of Medical Oncology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Hisanobu Oda
- Division of Integrative Medical Oncology, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Shuji Arita
- Department of Chemotherapy, Miyazaki Prefectural Miyazaki Hospital, Miyazaki, Japan
| | - Hiroki Taguchi
- Department of Gastroenterology, Saiseikai Sendai Hospital, Kagoshima, Japan
| | - Kengo Tsuneyoshi
- Department of Gastroenterology, Izumi General Medical Center, Kagoshima, Japan
| | - Yasunori Kawaguchi
- Department of Gastroenterology, Asakura Medical Association Hospital, Fukuoka, Japan
| | - Toshihiro Fujita
- Department of Gastroenterology, Saiseikai Sendai Hospital, Kagoshima, Japan
| | - Takahiro Sakae
- Department of Gastroenterology, Saiseikai Sendai Hospital, Kagoshima, Japan
| | - Kenta Nio
- Department of Medical Oncology, Sasebo Kyosai Hospital, Nagasaki, Japan
| | - Yasushi Ide
- Department of Internal Medicine, Karatsu Red Cross Hospital, Saga, Japan
| | - Norio Ureshino
- Department of Medical Oncology, Saga-Ken Medical Center Koseikan, Saga, Japan
| | - Tsuyoshi Shirakawa
- Clinical Hematology Oncology Treatment Study Group, Fukuoka, Japan
- Eikoh Hospital, Fukuoka, Japan
| | - Toshihiko Mizuta
- Department of Internal Medicine, Imari Arita Kyoritsu Hospital, Saga, Japan
| | - Kenji Mitsugi
- Department of Medical Oncology, Sasebo Kyosai Hospital, Nagasaki, Japan
- Department of Medical Oncology, Hamanomachi Hospital, Fukuoka, Japan
| |
Collapse
|
145
|
Wu J, Zhang Y, Wang H, Guo W, Li C, Yu Y, Liu H, Li F, Wang L, Xu J. Evaluating the benefits of adjuvant chemotherapy in patients with pancreatic cancer undergoing radical pancreatectomy after neoadjuvant therapy-a systematic review and meta-analysis. Front Oncol 2024; 14:1429386. [PMID: 39484040 PMCID: PMC11524795 DOI: 10.3389/fonc.2024.1429386] [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: 05/08/2024] [Accepted: 09/16/2024] [Indexed: 11/03/2024] Open
Abstract
Background More and more patients with pancreatic cancer (PC) received neoadjuvant therapy (NAT) and then underwent radical pancreatectomy. However, the benefit of adjuvant chemotherapy (AC) for these patients is still controversial. This study is designed to determine the benefits of postoperative AC for patients with PC undergoing NAT and radical resection. Methods We conducted a comprehensive search of the PubMed, Embase, Web of Science, and Cochrane Library databases, covering the period from their inception until 10 September 2023. Our analysis focused on the assessment of overall survival (OS) and recurrence-free survival (RFS) through meta-analysis. The fixed-effects model and the random-effects model were used to process the data. Hazard ratios (HRs) and 95% confidence intervals (95% CIs) were employed to determine the necessary of administering AC for patients with PC who have undergone NAT and radical resection. We retrieved 3,063 search results, of which 3,048 were excluded because of duplication or after applying our inclusion and exclusion criteria. Results A total of 15 studies with 21,113 patients (7,794 patients in the AC group and 13,319 in the non-AC group) were included, all of which reported OS, and three studies reported disease-free survival (DFS)/tumor-specific survival (CSS)/RFS. The final results showed that AC significantly improved OS and DFS/CSS/RFS in patients with PC who underwent pancreatectomy after NAT [OS: HR = 0.80, 95% CI (0.75∼0.86), P < 0.00001, I2 = 48%; DFS/CSS/RFS: HR = 0.53, 95% CI (0.41~0.69), P < 0.00001, I2 = 0%]. Furthermore, we performed subgroup analyses and demonstrated that AC provided a significant survival benefit for patients with PC after NAT and resection regardless of the tumor size [<2-cm subgroup: HR = 0.72, 95% CI (0.5∼0.94), P = 0.01; ≥2-cm subgroup: HR = 0.79, 95% CI (0.65∼0.96), P = 0.02] and the margin status [R0 subgroup: HR = 0.83, 95% CI (0.77∼0.88), P < 0.00001; R2 subgroup: HR = 0.75, 95% CI (0.61∼0.92), P = 0.007]. AC also benefited the patients with a stage N0 [HR = 0.79, 95% CI (0.74~0.84), P < 0.00001], N1 [HR = 0.78, 95% CI (0.72∼0.85), P < 0.00001], or poorly/undifferentiated tumor [HR = 0.76, 95% CI (0.66∼0.87), P < 0.0001] in survival but not in patients with a stage N2 [HR = 0.69, 95% CI (0.43∼1.09), P = 0.11] or well/moderately differentiated tumor [HR = 0.97, 95% CI (0.66∼1.42), P = 0.87]. Conclusions Although AC showed survival benefit for patients with PC undergoing radical pancreatectomy after NAT, we still need to consider the lymph node stage and the degree of differentiation of the tumor when we gave AC to a patient. High-quality prospective randomized controlled studies are required to well disclose the value of AC in patients with PC undergoing radical pancreatectomy after NAT. Systematic review registration https://www.crd.york.ac.uk/prospero/ PROSPERO, identifier CRD42023461365.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lei Wang
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Jianwei Xu
- *Correspondence: Jianwei Xu, ; Lei Wang,
| |
Collapse
|
146
|
Ouyang Y, Liu P, Chu L, Xiao Y, Zhu H, Qiang hao, Zhang C. Is chemotherapy beneficial? A retrospective study of chemotherapy in patients with invasive intraductal papillary-mucinous carcinoma. Heliyon 2024; 10:e38430. [PMID: 39430496 PMCID: PMC11489152 DOI: 10.1016/j.heliyon.2024.e38430] [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: 11/12/2023] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 10/22/2024] Open
Abstract
Background and aim Whether chemotherapy can improve the prognosis of invasive intraductal papillary-mucinous carcinoma (IPMC) still remains unclear. The aim of this study is to observe the difference in survival time of patients with invasive IPMC receiving or not receiving chemotherapy. Methods 117 patients with invasive IPMC were included in The Surveillance, Epidemiology, and End Results (SEER) database. These patients were subsequently divided into two subgroups according to whether they received chemotherapy or not: the non-chemotherapy group (patients who did not receivechemotherapy, N = 58), the chemotherapy group (patients who received chemotherapy, N = 59). The overall survival (OS) and cancer specific survival (CSS) of two treatment groups were evaluated. Results Before adjusting for pathology grade, the Kaplan-Meier analysis showed that the difference of survival time is not significant between non-chemotherapy group and chemotherapy group (P > 0.05), but the land-mark analysis showed that short-term death risk of the chemotherapy group is significantly lower than non-chemotherapy group (P < 0.05). After adjust the pathology grade, survival time of the chemotherapy group is significantly longer than non-chemotherapy group (P < 0.05). Univariate and multivariate Cox regression showed that chemotherapy was an independent prognostic protective factor for invasive IPMC (P < 0.05). Land-mark analysis showed that short-term death risk of the chemotherapy group is significantly lower than non-chemotherapy group in N1-N2 subgroup (P < 0.05). Conclusion Chemotherapy is an independent protective factor IPMC, especially reducing the risk of short-term death for IPMC patients with lymph node metastasis.
Collapse
Affiliation(s)
- Yonghao Ouyang
- Research Institute of General Surgery, Jinling Hospital, Nanjing 210000, China
| | - Pengpeng Liu
- Department of Hepatobiliary, Xuzhou Central Hospital, Xuzhou 221000, China
| | - Lihua Chu
- Jinggangshan University, Ji'an 343000,China
| | - Yi Xiao
- Research Institute of General Surgery, Jinling Hospital, Nanjing 210000, China
| | - Hong Zhu
- Department of Hepatobiliary, Xuzhou Central Hospital, Xuzhou 221000, China
| | - Qiang hao
- Department of Hepatobiliary, Xuzhou Central Hospital, Xuzhou 221000, China
| | - Caihua Zhang
- Department of Gastrointestinal, Xuzhou Central Hospital, Xuzhou 221000, China
| |
Collapse
|
147
|
Wang J, Yang J, Narang A, He J, Wolfgang C, Li K, Zheng L. Consensus, debate, and prospective on pancreatic cancer treatments. J Hematol Oncol 2024; 17:92. [PMID: 39390609 PMCID: PMC11468220 DOI: 10.1186/s13045-024-01613-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Pancreatic cancer remains one of the most aggressive solid tumors. As a systemic disease, despite the improvement of multi-modality treatment strategies, the prognosis of pancreatic cancer was not improved dramatically. For resectable or borderline resectable patients, the surgical strategy centered on improving R0 resection rate is consensus; however, the role of neoadjuvant therapy in resectable patients and the optimal neoadjuvant therapy of chemotherapy with or without radiotherapy in borderline resectable patients were debated. Postoperative adjuvant chemotherapy of gemcitabine/capecitabine or mFOLFIRINOX is recommended regardless of the margin status. Chemotherapy as the first-line treatment strategy for advanced or metastatic patients included FOLFIRINOX, gemcitabine/nab-paclitaxel, or NALIRIFOX regimens whereas 5-FU plus liposomal irinotecan was the only standard of care second-line therapy. Immunotherapy is an innovative therapy although anti-PD-1 antibody is currently the only agent approved by for MSI-H, dMMR, or TMB-high solid tumors, which represent a very small subset of pancreatic cancers. Combination strategies to increase the immunogenicity and to overcome the immunosuppressive tumor microenvironment may sensitize pancreatic cancer to immunotherapy. Targeted therapies represented by PARP and KRAS inhibitors are also under investigation, showing benefits in improving progression-free survival and objective response rate. This review discusses the current treatment modalities and highlights innovative therapies for pancreatic cancer.
Collapse
Affiliation(s)
- Junke Wang
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jie Yang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Amol Narang
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jin He
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Christopher Wolfgang
- Department of Surgery, New York University School of Medicine and NYU-Langone Medical Center, New York, NY, USA
| | - Keyu Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA.
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA.
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Multidisciplinary Gastrointestinal Cancer Laboratories Program, the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
148
|
Racu ML, Schiavo AA, Van Campenhout C, De Nève N, Masuy T, Maris C, Decaestecker C, Remmelink M, Salmon I, D'Haene N. Validation of a targeted next-generation sequencing panel for pancreatic ductal adenocarcinomas. Exp Mol Pathol 2024; 139:104920. [PMID: 39033589 DOI: 10.1016/j.yexmp.2024.104920] [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: 02/21/2024] [Revised: 07/07/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is reported to be amongst the cancers with the lowest survival rate at 5 years. In the present study we aimed to validate a targeted next-generation sequencing (tNGS) panel to use in clinical routine, investigating genes important for PDAC diagnostic, prognostic and potential theragnostic aspect. In this NGS panel we also designed target regions to inquire about loss of heterozygosity (LOH) of chromosome 18 that has been described to be possibly linked to a worse disease progression. Copy number alteration has also been explored for a subset of genes. The last two methods are not commonly used for routine diagnostic with tNGS panels and we investigated their possible contribution to better characterize PDAC. A series of 140 formalin-fixed paraffin-embedded (FFPE) PDAC samples from 140 patients was characterized using this panel. Ninety-two % of patients showed alterations in at least one of the investigated genes (most frequent KRAS, TP53, SMAD4, CDKN2A and RNF43). Regarding LOH evaluation, we were able to detect chr18 LOH starting at 20% cell tumor percentage. The presence of LOH on chr18 is associated with a worse disease- and metastasis-free survival, in uni- and multivariate analyses. The present study validates the use of a tNGS panel for PDAC characterization, also evaluating chr18 LOH status for prognostic stratification.
Collapse
Affiliation(s)
- Marie-Lucie Racu
- Department of Pathology, Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), CUB Hôpital Érasme, Route de Lennik, 808 1070 Brussels, Belgium
| | - Andrea Alex Schiavo
- Department of Pathology, Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), CUB Hôpital Érasme, Route de Lennik, 808 1070 Brussels, Belgium
| | - Claude Van Campenhout
- Department of Pathology, Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), CUB Hôpital Érasme, Route de Lennik, 808 1070 Brussels, Belgium
| | - Nancy De Nève
- Department of Pathology, Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), CUB Hôpital Érasme, Route de Lennik, 808 1070 Brussels, Belgium
| | - Thomas Masuy
- Department of Pathology, Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), CUB Hôpital Érasme, Route de Lennik, 808 1070 Brussels, Belgium
| | - Calliope Maris
- Department of Pathology, Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), CUB Hôpital Érasme, Route de Lennik, 808 1070 Brussels, Belgium
| | - Christine Decaestecker
- Digital Image Analysis in Pathology (DIAPath), Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium; Laboratory of Image Synthesis and Analysis (LISA), Brussels School of Engineering/École Polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Ixelles, Belgium
| | - Myriam Remmelink
- Department of Pathology, Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), CUB Hôpital Érasme, Route de Lennik, 808 1070 Brussels, Belgium
| | - Isabelle Salmon
- Department of Pathology, Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), CUB Hôpital Érasme, Route de Lennik, 808 1070 Brussels, Belgium
| | - Nicky D'Haene
- Department of Pathology, Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), CUB Hôpital Érasme, Route de Lennik, 808 1070 Brussels, Belgium.
| |
Collapse
|
149
|
Giuliante F, Panettieri E, Campisi A, Coppola A, Vellone M, De Rose AM, Ardito F. Treatment of oligo-metastatic pancreatic ductal adenocarcinoma to the liver: is there a role for surgery? A narrative review. Int J Surg 2024; 110:6163-6169. [PMID: 38818688 PMCID: PMC11486948 DOI: 10.1097/js9.0000000000001665] [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: 12/30/2023] [Accepted: 05/09/2024] [Indexed: 06/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a prognostically unfavorable malignancy that presents with distant metastases at the time of diagnosis in half of patients. Even if patients with metastatic PDAC have not been traditionally considered candidates for surgery, an increasing number of researchers have been investigating the efficacy of surgical treatment for patients with liver-only oligometastases from PDAC, showing promising results in extremely selected patients, mainly with metachronous metastases after perioperative chemotherapy. Nevertheless, a standardized definition of oligometastatic disease should be adopted and additional investigations focusing on the role of perioperative chemotherapy and tumor biology are warranted to reliably assess the role of resection for PDAC metastatic to the liver.
Collapse
Affiliation(s)
- Felice Giuliante
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| | - Elena Panettieri
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrea Campisi
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| | | | - Maria Vellone
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| | - Agostino M. De Rose
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| | - Francesco Ardito
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| |
Collapse
|
150
|
García García de Paredes A, Martínez Moneo E, Lariño-Noia J, Earl J. Pancreatic cancer screening in high-risk individuals. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2024; 116:519-522. [PMID: 39087662 DOI: 10.17235/reed.2024.10635/2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
The incidence of pancreatic cancer is increasing, although globally it represents less than 3% of all cancers. Despite advances in medical and surgical management, survival rates have not significantly improved in recent years. Consequently, pancreatic cancer, though relatively uncommon, is the third leading cause of cancer-related deaths. This is primarily due to the disease´s late detection. Symptoms appear late and are nonspecific, and over 80% of cases are diagnosed at an advanced stage and unsuitable for curative surgery, resulting in a five-year survival rate below 10%. However, the exceptional cases that are diagnosed early show five-year survival rates exceeding 80%. Therefore, one of the keys to improving pancreatic cancer prognosis lies in early detection, making screening in high-risk individuals a potentially crucial strategy.
Collapse
Affiliation(s)
| | | | | | - Julie Earl
- Biomarkers and Personalized Approach to Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)
| |
Collapse
|