1
|
Meng Y, Wang C, Usyk M, Kwak S, Peng C, Hu KS, Oberstein PE, Krogsgaard M, Li H, Hayes RB, Ahn J. Association of tumor microbiome with survival in resected early-stage PDAC. mSystems 2025; 10:e0122924. [PMID: 40013793 PMCID: PMC11915875 DOI: 10.1128/msystems.01229-24] [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: 09/11/2024] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
The pancreas tumor microbiota may influence tumor microenvironment and influence survival in early-stage pancreatic ductal adenocarcinoma (PDAC); however, current studies are limited and small. We investigated the relationship of tumor microbiota to survival in 201 surgically resected patients with localized PDAC (Stages I-II), from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) cohorts. We characterized the tumor microbiome using RNA-sequencing data. We examined the association of the tumor microbiome with overall survival (OS), via meta-analysis with the Cox PH model. A microbial risk score (MRS) was calculated from the OS-associated microbiota. We further explored whether the OS-associated microbiota is related to host tumor immune infiltration. PDAC tumor microbiome α- and β-diversities were not associated with OS; however, 11 bacterial species, including species of Gammaproteobacteria, confirmed by extensive resampling, were significantly associated with OS (all Q < 0.05). The MRS summarizing these bacteria was related to a threefold change in OS (hazard ratio = 2.96 per standard deviation change in the MRS, 95% confidence interval = 2.26-3.86). This result was consistent across the two cohorts and in stratified analyses by adjuvant therapy (chemotherapy/radiation). Identified microbiota and the MRS also exhibited association with memory B cells and naïve CD4+ T cells, which may be related to the immune landscape through BCR and TCR signaling pathways. Our study shows that a unique tumor microbiome structure, potentially affecting the tumor immune microenvironment, is associated with poorer survival in resected early-stage PDAC. These findings suggest that microbial mechanisms may be involved in PDAC survival, potentially informing PDAC prognosis and guiding personalized treatment strategies.IMPORTANCEMuch of the available data on the PDAC tumor microbiome and survival are derived from relatively small and heterogeneous studies, including those involving patients with advanced stages of pancreatic cancer. There is a critical knowledge gap in terms of the tumor microbiome and survival in early-stage patients treated by surgical resection; we expect that advancements in survival may initially be best achieved in these patients who are treated with curative intent.
Collapse
Affiliation(s)
- Yixuan Meng
- Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
| | - Chan Wang
- Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
| | - Mykhaylo Usyk
- Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
| | - Soyoung Kwak
- Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
| | - Chengwei Peng
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Kenneth S Hu
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Paul E Oberstein
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
| | - Michelle Krogsgaard
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
| | - Huilin Li
- Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
| | - Richard B Hayes
- Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
| | - Jiyoung Ahn
- Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
| |
Collapse
|
2
|
Al Zarkali O, Hussaini M, Kim DW, Yu HHM, Ott P, Ali S. A Rare Case of Simultaneous Brain and Leptomeningeal Metastases in a Patient With Pancreatic Cancer. Cureus 2025; 17:e79923. [PMID: 40171334 PMCID: PMC11961017 DOI: 10.7759/cureus.79923] [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] [Accepted: 03/02/2025] [Indexed: 04/03/2025] Open
Abstract
Pancreatic cancer rarely metastasizes to the brain or meninges. Here, we describe a case in which a patient developed both. Our patient, a 75-year-old male individual, was diagnosed with pancreatic tumor on computed tomography (CT) of the abdomen and pelvis after presenting for abdominal pain. The patient underwent an endoscopic ultrasound-guided fine needle aspiration of the pancreatic mass and pathology was consistent with adenocarcinoma of the pancreas. A positron emission tomography (PET) scan showed metabolically active pancreatic head mass, retroperitoneal soft tissue nodule/lymph node, soft tissue thickening adjacent to the celiac artery, and within the anterior left lung hilum. He was treated with neoadjuvant chemotherapy followed by Whipple procedure due to presence of oligometastasis with great response to initial treatment. He later developed a frontal headache while on surveillance. MRI of the brain demonstrated multiple intraparenchymal metastases as well as leptomeningeal carcinomatosis. The patient underwent a posterior fossa craniotomy for resection of the mass. The pathology was consistent with metastatic pancreatic adenocarcinoma. This case describes a malignancy that presented with a unique form of metastasis.
Collapse
Affiliation(s)
- Omar Al Zarkali
- Internal and Hospital Medicine, Moffitt Cancer Center, Tampa, USA
| | | | - Dae Won Kim
- Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, USA
| | | | - Paul Ott
- Internal and Hospital Medicine, Moffitt Cancer Center, Tampa, USA
| | - Sarah Ali
- Internal and Hospital Medicine, Moffitt Cancer Center, Tampa, USA
| |
Collapse
|
3
|
Lin Y, Liao Y, Shen J. Angiotensin system inhibitors improve survival in patients undergoing pancreatic cancer resection: a meta-analysis of real-world evidence. Expert Rev Anticancer Ther 2025:1-8. [PMID: 39910780 DOI: 10.1080/14737140.2025.2464208] [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: 09/25/2024] [Revised: 01/15/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND The role of angiotensin system inhibitors (ASIs) in modifying the prognosis for patients undergoing pancreatic cancer resection is not yet definitively established. This meta-analysis endeavors to consolidate existing real-world data to provide a robust, evidence-based assessment of their impact on clinical outcomes. METHODS A meticulous search strategy was devised and executed across PubMed, Embase, and Web of Science databases to retrieve all relevant studies evaluating the prognostic impact of ASIs in patients who have undergone resection for pancreatic cancer. Studies comparing survival outcomes between ASI users and non-users were included in the meta-analysis. Publication bias was assessed using funnel plotand Egger's test. Sensitivity analysis employing the leave-one-out approach was conducted to ensure the robustness and reliability of the pooled estimate. RESULTS Seven studies encompassing 8,549 patients were analyzed. The utilization of ASIs was significantly associated with improved overall survival (HR: 0.78; 95%CI: 0.68-0.89) in patients undergoing pancreatic cancer resection. Sensitivity analysis further validated the consistency and stability of the pooled result. CONCLUSION Current clinical evidence suggests that ASIs are associated with improved prognosis in patients who have undergone pancreatic cancer resection. These findings highlight the potential of ASIs as a beneficial adjunctive therapy in the management of resected pancreatic cancer, warranting their consideration in clinical management protocols. REGISTRATION PROSPERO (identifier: CRD42024580624).
Collapse
Affiliation(s)
- Yuxuan Lin
- Department of Pharmacy, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, Guangxi, PR China
| | - Yonghe Liao
- College of Pharmaceutical Science, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Jinhai Shen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, PR China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, Jiangsu, PR China
| |
Collapse
|
4
|
Borbély RZ, Teutsch B, Hegyi P. Incidence and Management of Splanchnic Vein Thrombosis in Pancreatic Diseases. United European Gastroenterol J 2025; 13:86-96. [PMID: 39743752 PMCID: PMC11866318 DOI: 10.1002/ueg2.12744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 01/04/2025] Open
Abstract
Splanchnic vein thrombosis (SVT) in pancreatic disease has a 20%-30% incidence rate, leading to increased mortality and complication rates. Therefore, the aim of this review is to summarize recent evidence about the incidence, risk factors, and management of pancreatic cancer, pancreatic cystic neoplasm-, and pancreatitis-related SVT. Doppler ultrasound should be the first imaging choice, followed by contrast-enhanced computed tomography or magnetic resonance imaging. Data regarding SVT treatment in acute pancreatitis and pancreatic cancer are scarce; however, for venous thromboembolism treatment, direct oral anticoagulants and low molecular weight heparin have been effective. Further trials must investigate the length of anticoagulant treatment and the need for interventional radiological procedures.
Collapse
Affiliation(s)
- Ruben Zsolt Borbély
- Centre for Translational MedicineSemmelweis UniversityBudapestHungary
- Department of Medical ImagingBajcsy‐Zsilinszky Hospital and ClinicBudapestHungary
| | - Brigitta Teutsch
- Centre for Translational MedicineSemmelweis UniversityBudapestHungary
- Institute for Translational MedicineMedical SchoolUniversity of PécsPécsHungary
- Department of RadiologyMedical Imaging CentreSemmelweis UniversityBudapestHungary
| | - Péter Hegyi
- Centre for Translational MedicineSemmelweis UniversityBudapestHungary
- Institute for Translational MedicineMedical SchoolUniversity of PécsPécsHungary
- Institute of Pancreatic DiseasesSemmelweis UniversityBudapestHungary
- Translational Pancreatology Research GroupInterdisciplinary Center of Excellence for Research Development and InnovationUniversity of SzegedSzegedHungary
| |
Collapse
|
5
|
Wehrle CJ, Chang J, Gross A, Perlmutter B, Naples R, Stackhouse K, Augustin T, Joyce D, Simon R, Schlegel A, Walsh RM, Naffouje SA, Parente A. Sequence of Chemotherapy May Not Impact Survival After Resection of Pancreatic Tail Adenocarcinoma. J Surg Oncol 2025. [DOI: 10.1002/jso.28086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 01/01/2025] [Indexed: 02/03/2025]
Abstract
ABSTRACTIntroductionPancreatic ductal adenocarcinoma (PDAC) of the body/tail is notably different than PDAC in the head of the pancreas. Surgery plus chemotherapy is known to improve outcomes for all PDAC. The sequence of this therapy is well studied in head cancers yet has never been evaluated systematically in relation to distal pancreatectomy (DP).MethodsPatients receiving DP for PDAC and who received chemotherapy were included. Patients were compared receiving neoadjuvant systemic therapy (NAST) only, adjuvant (AST) only, both NAST + AST, and who received total neoadjuvant therapy (TNT), defined as > 24 weeks NAST before DP. PSM was performed 1:1 between AST and each other group creating quadruplets of patients for analysis. Matching factors were determined by multivariate cox‐regression analysis of factors independently affecting survival. Survival was considered from diagnosis and from surgery to account for potential biases.ResultsIn total, 4677 patients were selected with 400 (8.6%) receiving TNT, 536 (11.5%) NAST, 3235 (69.2%) AST, and 506 (10.8%) NAST + AST. A total of 341 quadruplets were selected after PSM. There were no differences in comorbidities, T/N‐stage, retrieved or positive lymph nodes, and margin status after matching. Kaplan–Meier analysis showed no difference in median OS between the matched treatment groups (33.71 ± 2.07 vs. 35.22 ± 1.62 vs. 32.53 ± 3.31 vs. 37.88 ± 1.90, respectively; log‐rank p = 0.464). Five‐year OS was not different between the groups (21% vs. 18% vs. 20% vs. 25%, respectively; p = 0.501).ConclusionThe sequence of chemotherapy and surgery did not impact survival in distal PDAC. Providers should tailor an individualized approach designed to maximize the chance of completing both treatments.
Collapse
Affiliation(s)
- Chase J. Wehrle
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Jenny Chang
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Abby Gross
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Breanna Perlmutter
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Robert Naples
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | | | - Toms Augustin
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Daniel Joyce
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Robert Simon
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Andrea Schlegel
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - R. Matthew Walsh
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Samer A. Naffouje
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Alessandro Parente
- Institute of Liver Studies, King's College Hospital NHS Foundation Trust London UK
| |
Collapse
|
6
|
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
|
7
|
Jeremić S, Avdović E, Dolićanin Z, Vojinović R, Antonijević M, Marković Z. In silico study of novel coumarin derivatives as potential agents in the pancreatic cancer treatment. Comput Methods Biomech Biomed Engin 2024:1-15. [PMID: 39568331 DOI: 10.1080/10255842.2024.2431345] [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: 10/12/2023] [Revised: 11/01/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest diseases. Here are investigated two synthesized and two hypothetical coumarin derivatives, and their capacity to be used in the PDAC targeted treatment. The inhibitory activity of these four molecules against PARP, ATM, and CHK1 proteins responsible for DNA molecule repair was examined by docking and molecular dynamic analysis. ADMET analysis was applied to determine the pharmacokinetic properties of the tested compounds. The applied theoretical approach showed that the biomedical activity of the investigated coumarins is comparable to the inhibitory activity and pharmacokinetic properties of Olaparib, already used in the PDAC treatment.
Collapse
Affiliation(s)
- Svetlana Jeremić
- Department of Natural Science and Mathematics, State University of Novi Pazar, Novi Pazar, Serbia
| | - Edina Avdović
- Institute for Information Technologies, Kragujevac, University of Kragujevac, Kragujevac, Serbia
| | - Zana Dolićanin
- Department of Natural Science and Mathematics, State University of Novi Pazar, Novi Pazar, Serbia
| | - Radiša Vojinović
- Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Marko Antonijević
- Institute for Information Technologies, Kragujevac, University of Kragujevac, Kragujevac, Serbia
| | - Zoran Marković
- Department of Natural Science and Mathematics, State University of Novi Pazar, Novi Pazar, Serbia
- Institute for Information Technologies, Kragujevac, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
8
|
Hoffman D, Ganjouei AA, Hernandez FR, Ifuku K, Miller P, Glencer A, Corvera C, Kirkwood K, Alseidi A, Adam M, Maker A, Hirose K, Hirose R, Nakakura EK. Graft choice in pancreatectomy with vascular resection: equivalent safety in selected patients. J Gastrointest Surg 2024; 28:1799-1804. [PMID: 39181231 DOI: 10.1016/j.gassur.2024.08.016] [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: 05/04/2024] [Revised: 06/17/2024] [Accepted: 08/17/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Pancreatic cancer is the third leading cause of cancer-related death in the United States, with surgical resection being the only option for long-term survival. The ability to manage vascular involvement has expanded the pool of patients who are able to undergo resection with curative intent. However, not all vascular involvements can be detected preoperatively. This study aimed to investigate the patterns of vascular resection and methods of repair or reconstruction METHODS: This was a single-center retrospective review of adult patients undergoing pancreatectomy with vascular involvement at a tertiary care referral hospital between 2010 and 2022. The primary endpoint was graft thrombosis within 90 days. RESULTS A total of 147 patients were included in the study. Of note, 21.8% of patients were not suspected of having vascular involvement preoperatively. Moreover, 68.0% of patients required vascular reconstruction, whereas the remaining 32.0% of patients underwent repair (either primary repair or patch angioplasty). Most patients who underwent reconstruction underwent primary end-to-end anastomosis (63.0%), with 19 patients requiring autologous interposition grafts and 16 patients requiring CryoVein interposition grafts. Univariate analysis found no clinical or technical predictors of early or 90-day thrombosis, including graft choice. In addition, 30- and 90-day mortalities occurred in 1 and 7 patients, respectively. CONCLUSION Pancreatectomy with vascular resection can be performed with low mortality in carefully selected patients. Unsuspected vascular involvement is relatively common (1 in 5). If autologous graft is not readily available, CryoVein is a safe alternative with similar perioperative outcomes.
Collapse
Affiliation(s)
- Daniel Hoffman
- Department of Surgery, University of California, San Francisco, CA, United States
| | - Amir Ashraf Ganjouei
- Department of Surgery, University of California, San Francisco, CA, United States
| | | | - Kelli Ifuku
- Department of Surgery, University of California, San Francisco, CA, United States
| | - Phoebe Miller
- Department of Surgery, University of California, San Francisco, CA, United States
| | - Alexa Glencer
- Department of Surgery, University of California, San Francisco, CA, United States
| | - Carlos Corvera
- Department of Surgery, University of California, San Francisco, CA, United States; Section of Hepatopancreaticobiliary Surgery, Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, United States
| | - Kimberly Kirkwood
- Department of Surgery, University of California, San Francisco, CA, United States; Section of Hepatopancreaticobiliary Surgery, Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, United States
| | - Adnan Alseidi
- Department of Surgery, University of California, San Francisco, CA, United States; Section of Hepatopancreaticobiliary Surgery, Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, United States
| | - Mohamed Adam
- Department of Surgery, University of California, San Francisco, CA, United States; Section of Hepatopancreaticobiliary Surgery, Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, United States
| | - Ajay Maker
- Department of Surgery, University of California, San Francisco, CA, United States
| | - Kenzo Hirose
- Department of Surgery, University of California, San Francisco, CA, United States; UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States
| | - Ryutaro Hirose
- Department of Surgery, University of California, San Francisco, CA, United States; UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States; Division of Transplant Surgery, Department of Surgery, University of California, San Francisco, CA, United States
| | - Eric K Nakakura
- Department of Surgery, University of California, San Francisco, CA, United States; Section of Hepatopancreaticobiliary Surgery, Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, CA, United States; UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States.
| |
Collapse
|
9
|
Liao H, Yuan J, Liu C, Zhang J, Yang Y, Liang H, Liu H, Chen S, Li Y. One novel transfer learning-based CLIP model combined with self-attention mechanism for differentiating the tumor-stroma ratio in pancreatic ductal adenocarcinoma. LA RADIOLOGIA MEDICA 2024; 129:1559-1574. [PMID: 39412688 DOI: 10.1007/s11547-024-01902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/05/2024] [Indexed: 11/12/2024]
Abstract
PURPOSE To develop a contrastive language-image pretraining (CLIP) model based on transfer learning and combined with self-attention mechanism to predict the tumor-stroma ratio (TSR) in pancreatic ductal adenocarcinoma on preoperative enhanced CT images, in order to understand the biological characteristics of tumors for risk stratification and guiding feature fusion during artificial intelligence-based model representation. MATERIAL AND METHODS This retrospective study collected a total of 207 PDAC patients from three hospitals. TSR assessments were performed on surgical specimens by pathologists and divided into high TSR and low TSR groups. This study developed one novel CLIP-adapter model that integrates the CLIP paradigm with a self-attention mechanism for better utilizing features from multi-phase imaging, thereby enhancing the accuracy and reliability of tumor-stroma ratio predictions. Additionally, clinical variables, traditional radiomics model and deep learning models (ResNet50, ResNet101, ViT_Base_32, ViT_Base_16) were constructed for comparison. RESULTS The models showed significant efficacy in predicting TSR in PDAC. The performance of the CLIP-adapter model based on multi-phase feature fusion was superior to that based on any single phase (arterial or venous phase). The CLIP-adapter model outperformed traditional radiomics models and deep learning models, with CLIP-adapter_ViT_Base_32 performing the best, achieving the highest AUC (0.978) and accuracy (0.921) in the test set. Kaplan-Meier survival analysis showed longer overall survival in patients with low TSR compared to those with high TSR. CONCLUSION The CLIP-adapter model designed in this study provides a safe and accurate method for predicting the TSR in PDAC. The feature fusion module based on multi-modal (image and text) and multi-phase (arterial and venous phase) significantly improves model performance.
Collapse
Affiliation(s)
- Hongfan Liao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiang Yuan
- College of Computer and Information Science, Southwest University, Chongqing, 400715, China
| | - Chunhua Liu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Jiao Zhang
- Department of Radiology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaying Yang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Hongwei Liang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Haotian Liu
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Shanxiong Chen
- College of Computer and Information Science, Southwest University, Chongqing, 400715, China.
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
10
|
Seufferlein T, Mayerle J, Boeck S, Brunner T, Ettrich TJ, Grenacher L, Gress TM, Hackert T, Heinemann V, Kestler A, Sinn M, Tannapfel A, Wedding U, Uhl W. S3-Leitlinie Exokrines Pankreaskarzinom – Version 3.1. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:e874-e995. [PMID: 39389103 DOI: 10.1055/a-2338-3533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Affiliation(s)
| | | | | | - Thomas Brunner
- Universitätsklinik für Strahlentherapie-Radioonkologie, Medizinische Universität Graz, Austria
| | | | | | - Thomas Mathias Gress
- Gastroenterologie und Endokrinologie Universitätsklinikum Gießen und Marburg, Germany
| | - Thilo Hackert
- Klinik und Poliklinik für Allgemein-, Viszeral- und Thoraxchirurgie, Universitätsklinikum Hamburg-Eppendorf, Germany
| | - Volker Heinemann
- Medizinische Klinik und Poliklinik III, Klinikum der Universität München-Campus Grosshadern, München, Germany
| | | | - Marianne Sinn
- Medizinische Klinik und Poliklinik II Onkologie und Hämatologie, Universitätsklinikum Hamburg-Eppendorf, Germany
| | | | | | - Waldemar Uhl
- Allgemein- und Viszeralchirurgie, St Josef-Hospital, Bochum, Germany
| |
Collapse
|
11
|
Lu PW, Lyu HG, Prakash LR, Chiang YJS, Maxwell JE, Snyder RA, Kim MP, Tzeng CWD, Katz MHG, Ikoma N. Effect of surgical approach on early return to intended oncologic therapy after resection for pancreatic ductal adenocarcinoma. Surg Endosc 2024; 38:4986-4995. [PMID: 38987482 DOI: 10.1007/s00464-024-11022-3] [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: 03/08/2024] [Accepted: 06/30/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Although robotic pancreatectomy may facilitate an earlier functional recovery, the impact of a robotic pancreatectomy program during its early experience on the timing of return to intended oncologic therapy (RIOT) after surgery is unknown. METHODS In this retrospective cohort study, we used propensity score matching with a 1:2 ratio to compare patients who underwent robotic or open surgery (distal pancreatectomy or pancreatoduodenectomy) for pancreatic ductal adenocarcinoma (PDAC) during the first 3 years of our robotic pancreatectomy experience (January 2018-December 2021). Generalized estimating equations modeling was used to evaluate the effect of surgical approach on early RIOT, defined as adjuvant chemotherapy initiation within 8 weeks after surgery, and late RIOT, defined as initiation within 12 weeks after surgery. RESULTS The matched cohort included 26 patients who underwent robotic pancreatectomy and 52 patients who underwent open pancreatectomy. Rates of receipt of adjuvant chemotherapy were 96.2% and 78.9%, respectively. Rate of early RIOT in the robotic group (73.1% was higher than that in the open group (44.2%; P = 0.018). In multivariable analysis, a robotic approach was associated with early RIOT (odds ratio, 3.54; 95% confidence interval 1.08-11.62; P = 0.038). Surgical approach did not impact late RIOT (odds ratio, 3.21; 95% confidence interval 0.71-14.38; P = 0.128). CONCLUSIONS Compared with open pancreatectomy, robotic pancreatectomy did not delay RIOT. In fact, odds of early RIOT were increased, which supports the oncological safety of our robotic pancreatectomy program during its implementation.
Collapse
Affiliation(s)
- Pamela W Lu
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Heather G Lyu
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Laura R Prakash
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Yi-Ju Sabrina Chiang
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Jessica E Maxwell
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Rebecca A Snyder
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Michael P Kim
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Ching-Wei D Tzeng
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Matthew H G Katz
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Naruhiko Ikoma
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
12
|
Carpenter EL, Van Decar SG, McCarthy PM, Valdera FA, Adams AM, O'Shea AE, Smolinsky T, Thomas K, Clifton GT, Newhook TE, Peoples GE, Nelson DW, Vreeland TJ. The benefit of adjuvant chemotherapy following pancreaticoduodenectomy for pancreatic adenocarcinoma depends on response to neoadjuvant therapy. J Surg Oncol 2024; 130:109-116. [PMID: 38801055 DOI: 10.1002/jso.27689] [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: 01/31/2024] [Revised: 04/15/2024] [Accepted: 05/11/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND The benefit of adjuvant therapy (AT) remains unclear in pancreatic ductal adenocarcinoma (PDAC) after neoadjuvant therapy (NAT) and surgical resection. METHODS The 2019 National Cancer Database was queried for patients with non-metastatic PDAC who received NAT followed by pancreaticoduodenectomy. Only patients with data regarding receipt of AT were included. Patients were classified if they had nodal down-staging specifically, or any downstaging (Tumor, Nodal, or overall). Propensity score matching (PSM) adjusted for pretreatment covariate imbalance between groups. The weighted Kaplan-Meier method and log-rank test were used to estimate the cumulative survival. RESULTS After exclusion criteria and PSM, a total of 2784 patients remained; 1689 (60.7%) received AT and 1095 (39.3%) did not receive AT. Among all, those with additional AT had a significantly improved overall survival (OS) (p < 0.001). Upon evaluation of patients without downstaging after NAT, those who received AT had improved OS (no nodal downstaging or any downstaging; p = 0.002; p = 0.001). When evaluating patients with downstaging after NAT, those receiving AT did not have improved OS (nodal downstaging or any downstaging: p = 0.352; p = 0.99). CONCLUSION Response to NAT appears to correlate with the benefit of AT following pancreaticoduodenectomy; patients who have a favorable response to NAT may not benefit from AT.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Timothy E Newhook
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | |
Collapse
|
13
|
Daamen LA, van Goor IWJM, Groot VP, Andel PCM, Brosens LAA, Busch OR, Cirkel GA, Mohammad NH, Heerkens HD, de Hingh IHJT, Hoogwater F, van Laarhoven HWM, Los M, Meijer GJ, de Meijer VE, Pande R, Roberts KJ, Stoker J, Stommel MWJ, van Tienhoven G, Verdonk RC, Verkooijen HM, Wessels FJ, Wilmink JW, Besselink MG, van Santvoort HC, Intven MPW, Molenaar IQ. Recurrent disease detection after resection of pancreatic ductal adenocarcinoma using a recurrence-focused surveillance strategy (RADAR-PANC): protocol of an international randomized controlled trial according to the Trials within Cohorts design. Trials 2024; 25:401. [PMID: 38902836 PMCID: PMC11188210 DOI: 10.1186/s13063-024-08223-5] [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: 03/20/2024] [Accepted: 06/03/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Disease recurrence remains one of the biggest concerns in patients after resection of pancreatic ductal adenocarcinoma (PDAC). Despite (neo)adjuvant systemic therapy, most patients experience local and/or distant PDAC recurrence within 2 years. High-level evidence regarding the benefits of recurrence-focused surveillance after PDAC resection is missing, and the impact of early detection and treatment of recurrence on survival and quality of life is unknown. In most European countries, recurrence-focused follow-up after surgery for PDAC is currently lacking. Consequently, guidelines regarding postoperative surveillance are based on expert opinion and other low-level evidence. The recent emergence of more potent local and systemic treatment options for PDAC recurrence has increased interest in early diagnosis. To determine whether early detection and treatment of recurrence can lead to improved survival and quality of life, we designed an international randomized trial. METHODS This randomized controlled trial is nested within an existing prospective cohort in pancreatic cancer centers in the Netherlands (Dutch Pancreatic Cancer Project; PACAP) and the United Kingdom (UK) (Pancreas Cancer: Observations of Practice and survival; PACOPS) according to the "Trials within Cohorts" (TwiCs) design. All PACAP/PACOPS participants with a macroscopically radical resection (R0-R1) of histologically confirmed PDAC, who provided informed consent for TwiCs and participation in quality of life questionnaires, are included. Participants randomized to the intervention arm are offered recurrence-focused surveillance, existing of clinical evaluation, serum cancer antigen (CA) 19-9 testing, and contrast-enhanced computed tomography (CT) of chest and abdomen every three months during the first 2 years after surgery. Participants in the control arm of the study will undergo non-standardized clinical follow-up, generally consisting of clinical follow-up with imaging and serum tumor marker testing only in case of onset of symptoms, according to local practice in the participating hospital. The primary endpoint is overall survival. Secondary endpoints include quality of life, patterns of recurrence, compliance to and costs of recurrence-focused follow-up, and the impact on recurrence-focused treatment. DISCUSSION The RADAR-PANC trial will be the first randomized controlled trial to generate high level evidence for the current clinical equipoise regarding the value of recurrence-focused postoperative surveillance with serial tumor marker testing and routine imaging in patients after PDAC resection. The Trials within Cohort design allows us to study the acceptability of recurrence-focused surveillance among cohort participants and increases the generalizability of findings to the general population. While it is strongly encouraged to offer all trial participants treatment at time of recurrence diagnosis, type and timing of treatment will be determined through shared decision-making. This might reduce the potential survival benefits of recurrence-focused surveillance, although insights into the impact on patients' quality of life will be obtained. TRIAL REGISTRATION Clinicaltrials.gov, NCT04875325 . Registered on May 6, 2021.
Collapse
Affiliation(s)
- L A Daamen
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands
- Division of Imaging, UMC Utrecht Cancer Center, Utrecht University, Utrecht, the Netherlands
| | - I W J M van Goor
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands.
- Department of Radiation Oncology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands.
| | - V P Groot
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands
| | - P C M Andel
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands
| | - L A A Brosens
- Department of Pathology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - O R Busch
- Amsterdam UMC, Department of Surgery, Location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - G A Cirkel
- Department of Medical Oncology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, St. Antonius Hospital Nieuwegein & Meander Medical Center, Utrecht University, Utrecht, the Netherlands
| | - N Haj Mohammad
- Department of Medical Oncology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, St. Antonius Hospital Nieuwegein & Meander Medical Center, Utrecht University, Utrecht, the Netherlands
| | - H D Heerkens
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - I H J T de Hingh
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands
| | - F Hoogwater
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - H W M van Laarhoven
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC, Department of Medical Oncology, Location University of Amsterdam, Amsterdam, the Netherlands
| | - M Los
- Department of Medical Oncology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, St. Antonius Hospital Nieuwegein & Meander Medical Center, Utrecht University, Utrecht, the Netherlands
| | - G J Meijer
- Department of Radiation Oncology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands
| | - V E de Meijer
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - R Pande
- Department of Hepatopancreatobiliary Surgery and Liver Transplantation, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, UK
| | - K J Roberts
- Department of Hepatopancreatobiliary Surgery and Liver Transplantation, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, UK
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - J Stoker
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC, Department of Radiology, Location University of Amsterdam, Amsterdam, the Netherlands
| | - M W J Stommel
- Department of Surgery, Radboud University Medical Center, Nijmegen, the Netherlands
| | - G van Tienhoven
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC, Department of Radiation Oncology, Location University of Amsterdam, Amsterdam, the Netherlands
| | - R C Verdonk
- Department of Gastroenterology and Hepatology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands
| | - H M Verkooijen
- Division of Imaging, UMC Utrecht Cancer Center, Utrecht University, Utrecht, the Netherlands
| | - F J Wessels
- Department of Radiology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands
| | - J W Wilmink
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC, Department of Medical Oncology, Location University of Amsterdam, Amsterdam, the Netherlands
| | - M G Besselink
- Amsterdam UMC, Department of Surgery, Location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - H C van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands
| | - M P W Intven
- Department of Radiation Oncology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands
| | - I Q Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands
| |
Collapse
|
14
|
Goetze TO, Reichart A, Bankstahl US, Pauligk C, Loose M, Kraus TW, Elshafei M, Bechstein WO, Trojan J, Behrend M, Homann N, Venerito M, Bohle W, Varvenne M, Bolling C, Behringer DM, Kratz-Albers K, Siegler GM, Hozaeel W, Al-Batran SE. Adjuvant Gemcitabine Versus Neoadjuvant/Adjuvant FOLFIRINOX in Resectable Pancreatic Cancer: The Randomized Multicenter Phase II NEPAFOX Trial. Ann Surg Oncol 2024; 31:4073-4083. [PMID: 38459418 PMCID: PMC11076394 DOI: 10.1245/s10434-024-15011-7] [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: 08/31/2023] [Accepted: 01/21/2024] [Indexed: 03/10/2024]
Abstract
BACKGROUND Although addition of adjuvant chemotherapy is the current standard, the prognosis of pancreatic cancers still remains poor. The NEPAFOX trial evaluated perioperative treatment with FOLFIRINOX in resectable pancreatic cancer. PATIENTS AND METHODS This multicenter phase II trial randomized patients with resectable or borderline resectable pancreatic cancer without metastases into arm (A,) upfront surgery plus adjuvant gemcitabine, or arm (B,) perioperative FOLFIRINOX. The primary endpoint was overall survival (OS). RESULTS Owing to poor accrual, recruitment was prematurely stopped after randomization of 40 of the planned 126 patients (A: 21, B: 19). Overall, approximately three-quarters were classified as primarily resectable (A: 16, B: 15), and the remaining patients were classified as borderline resectable (A: 5, B: 4). Of the 12 evaluable patients, 3 achieved partial response under neoadjuvant FOLFIRINOX. Of the 21 patients in arm A and 19 patients in arm B, 17 and 7 underwent curative surgery, and R0-resection was achieved in 77% and 71%, respectively. Perioperative morbidity occurred in 72% in arm A and 46% in arm B, whereas non-surgical toxicity was comparable in both arms. Median RFS/PFS was almost doubled in arm B (14.1 months) compared with arm A (8.4 months) in the population with surgical resection, whereas median OS was comparable between both arms. CONCLUSIONS Although the analysis was only descriptive owing to small patient numbers, no safety issues regarding surgical complications were observed in the perioperative FOLFIRINOX arm. Thus, considering the small number of patients, perioperative treatment approach appears feasible and potentially effective in well-selected cohorts of patients. In pancreatic cancer, patient selection before initiation of neoadjuvant therapy appears to be critical.
Collapse
Affiliation(s)
- Thorsten O Goetze
- Krankenhaus Nordwest, Institut für Klinisch Onkologische Forschchung IKF, University Cancer Center (UCT) Frankfurt, Frankfurt, Germany.
- University Cancer Center (UCT) Frankfurt, Goethe Universität, Frankfurt, Germany.
- Frankfurter Institut für Klinische Krebsforschung IKF am Krankenhaus Nordwest, Frankfurt, Germany.
| | - Alexander Reichart
- Krankenhaus Nordwest, Institut für Klinisch Onkologische Forschchung IKF, University Cancer Center (UCT) Frankfurt, Frankfurt, Germany
| | - Ulli S Bankstahl
- Krankenhaus Nordwest, Institut für Klinisch Onkologische Forschchung IKF, University Cancer Center (UCT) Frankfurt, Frankfurt, Germany
| | - Claudia Pauligk
- Frankfurter Institut für Klinische Krebsforschung IKF am Krankenhaus Nordwest, Frankfurt, Germany
| | - Maria Loose
- Frankfurter Institut für Klinische Krebsforschung IKF am Krankenhaus Nordwest, Frankfurt, Germany
| | - Thomas W Kraus
- Krankenhaus Nordwest, Allgemein-, Viszeral- und Minimal Invasive Chirurgie, Frankfurt, Germany
| | - Moustafa Elshafei
- Krankenhaus Nordwest, Allgemein-, Viszeral- und Minimal Invasive Chirurgie, Frankfurt, Germany
| | - Wolf O Bechstein
- Klinik für Allgemein-, Viszeral-, Transplantations- und Thoraxchirurgie, Universitätsklinikum Frankfurt, Frankfurt, Germany
| | - Jörg Trojan
- Gastrointestinale Onkologie, Universitätsklinikum Frankfurt, Frankfurt, Germany
| | - Matthias Behrend
- Viszeral-, Thorax- und Gefäßchirurgie, DONAUISAR Klinikum Deggendorf, Deggendorf, Germany
| | - Nils Homann
- Medizinische Klinik II, Klinikum Wolfsburg, Wolfsburg, Germany
| | - Marino Venerito
- Klinik für Gastroenterologie, Hepatologie und Infektiologie, Universitätsklinikum Magdeburg, Magdeburg, Germany
| | - Wolfram Bohle
- Klinik für Gastroenterologie, Gastroenterologische Onkologie, Klinikum Stuttgart, Stuttgart, Germany
- Hepatologie, Infektiologie und Pneumologie, Stuttgart, Germany
| | | | - Claus Bolling
- Hämatologie/Onkologie, Agaplesion Markus Krankenhaus, Frankfurt, Germany
| | - Dirk M Behringer
- Klinik für Hämatologie, Onkologie und Palliativmedizin, Augusta-Kranken-Anstalt Bochum, Bochum, Germany
| | | | - Gabriele M Siegler
- Klinikum Nürnberg Nord/Paracelsus Medizinische Privatuniversität, Medizinische Klinik, Hämatologie/Onkologie, Nürnberg, Germany
| | - Wael Hozaeel
- Krankenhaus Nordwest, Institut für Klinisch Onkologische Forschchung IKF, University Cancer Center (UCT) Frankfurt, Frankfurt, Germany
| | - Salah-Eddin Al-Batran
- Krankenhaus Nordwest, Institut für Klinisch Onkologische Forschchung IKF, University Cancer Center (UCT) Frankfurt, Frankfurt, Germany
- University Cancer Center (UCT) Frankfurt, Goethe Universität, Frankfurt, Germany
- Frankfurter Institut für Klinische Krebsforschung IKF am Krankenhaus Nordwest, Frankfurt, Germany
| |
Collapse
|
15
|
He J, Lv N, Yang Z, Luo Y, Zhong W, Wu C. Comparing upfront surgery with neoadjuvant treatments in patients with resectable, borderline resectable or locally advanced pancreatic cancer: a systematic review and network meta-analysis of randomized clinical trials. Int J Surg 2024; 110:3900-3909. [PMID: 38935819 PMCID: PMC11175811 DOI: 10.1097/js9.0000000000001313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/25/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND The aim was to explore the optimal neoadjuvant therapy strategy for resectable, borderline resectable, and locally advanced pancreatic cancer, in order to provide a theoretical basis for the development of new neoadjuvant treatment protocols for clinical use. PATIENTS AND METHODS The authors reviewed literature titles and abstracts comparing three treatment strategies (neoadjuvant chemoradiotherapy, neoadjuvant chemotherapy, and upfront surgery) in PubMed, Embase, The Cochrane Library, Web of Science from 2009 to 2023 to estimate relative odds ratios for resection rate and hazard ratios (HRs) for overall survival (OS) in all include trials. RESULTS A total of nine studies involving 889 patients were included in the analysis. The treatment methods included upfront surgery, neoadjuvant chemotherapy, and neoadjuvant chemoradiotherapy followed by surgery. The network meta-analysis results demonstrated that neoadjuvant chemoradiotherapy followed by surgery was an effective approach in improving OS for resectable and borderline resectable pancreatic cancer (RPC) patients compared to upfront surgery (HR: 0.79, 95% CI: 0.64-0.98) and neoadjuvant chemotherapy (HR: 0.79, 95% CI: 0.64-0.98). Additionally, neoadjuvant chemoradiotherapy significantly increased the margin negative resection (R0) rate and pathological negative lymph node (pN0) rate in patients with resectable and borderline RPC. However, it is worth noting that neoadjuvant chemoradiotherapy increased the risk of grade 3 or higher treatment-related adverse events, including in patients with locally advanced pancreatic cancer. CONCLUSIONS The current evidence suggests that neoadjuvant chemoradiotherapy followed by surgery is the optimal choice for treating patients with resectable and borderline RPC. Future research should focus on optimizing neoadjuvant chemoradiotherapy regimens to effectively improve OS while reducing the occurrence of adverse events.
Collapse
Affiliation(s)
| | | | | | | | | | - Chunli Wu
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Liaoning, China
| |
Collapse
|
16
|
Bilreiro C, Andrade L, Santiago I, Marques RM, Matos C. Imaging of pancreatic ductal adenocarcinoma - An update for all stages of patient management. Eur J Radiol Open 2024; 12:100553. [PMID: 38357385 PMCID: PMC10864763 DOI: 10.1016/j.ejro.2024.100553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/16/2024] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is a common and lethal cancer. From diagnosis to disease staging, response to neoadjuvant therapy assessment and patient surveillance after resection, imaging plays a central role, guiding the multidisciplinary team in decision-planning. Review aims and findings This review discusses the most up-to-date imaging recommendations, typical and atypical findings, and issues related to each step of patient management. Example cases for each relevant condition are presented, and a structured report for disease staging is suggested. Conclusion Despite current issues in PDAC imaging at different stages of patient management, the radiologist is essential in the multidisciplinary team, as the conveyor of relevant imaging findings crucial for patient care.
Collapse
Affiliation(s)
- Carlos Bilreiro
- Radiology Department, Champalimaud Foundation, Lisbon, Portugal
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
- Nova Medical School, Lisbon, Portugal
| | - Luísa Andrade
- Radiology Department, Champalimaud Foundation, Lisbon, Portugal
| | - Inês Santiago
- Radiology Department, Champalimaud Foundation, Lisbon, Portugal
| | - Rui Mateus Marques
- Nova Medical School, Lisbon, Portugal
- Radiology Department, Hospital de S. José, Lisbon, Portugal
| | - Celso Matos
- Radiology Department, Champalimaud Foundation, Lisbon, Portugal
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| |
Collapse
|
17
|
Manojlovic N, Savic G, Manojlovic S. Neoadjuvant treatment of pancreatic ductal adenocarcinoma: Whom, when and how. World J Gastrointest Surg 2024; 16:1223-1230. [PMID: 38817288 PMCID: PMC11135299 DOI: 10.4240/wjgs.v16.i5.1223] [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: 12/28/2023] [Revised: 03/13/2024] [Accepted: 04/22/2024] [Indexed: 05/23/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), which is notorious for its aggressiveness and poor prognosis, remains an area of great unmet medical need, with a 5-year survival rate of 10% - the lowest of all solid tumours. At diagnosis, only 20% of patients have resectable pancreatic cancer (RPC) or borderline RPC (BRPC) disease, while 80% of patients have unresectable tumours that are locally advanced pancreatic cancer (LAPC) or have distant metastases. Nearly 60% of patients who undergo upfront surgery for RPC are unable to receive adequate adjuvant chemotherapy (CHT) because of postoperative complications and early cancer recurrence. An important paradigm shift to achieve better outcomes has been the sequence of therapy, with neoadjuvant CHT preceding surgery. Three surgical stages have emerged for the preoperative assessment of nonmetastatic pancreatic cancers: RPC, BRPC, and LAPC. The main goal of neoadjuvant treatment (NAT) is to improve postoperative outcomes through enhanced selection of candidates for curative-intent surgery by identifying patients with aggressive or metastatic disease during initial CHT, reducing tumour volume before surgery to improve the rate of margin-negative resection (R0 resection, a microscopic margin-negative resection), reducing the rate of positive lymph node occurrence at surgery, providing early treatment of occult micrometastatic disease, and assessing tumour chemosensitivity and tolerance to treatment as potential surgical criteria. In this editorial, we summarize evidence concerning NAT of PDAC, providing insights into future practice and study design. Future research is needed to establish predictive biomarkers, measures of therapeutic response, and multidisciplinary strategies to improve patient-centered outcomes.
Collapse
Affiliation(s)
- Nebojsa Manojlovic
- Clinic for Gastroenterology and Hepatology, Military Medical Academy, Faculty of Medicine of the Military Medical Academy, University of Defence, Belgrade 11000, Serbia
| | - Goran Savic
- Military Medical Academy, Faculty of Medicine of the Military Medical Academy, University of Defence, Belgrade 11000, Serbia
| | | |
Collapse
|
18
|
Yu HJ, Jang E, Woo A, Han IW, Jeon HG, Linh VTN, Park SG, Jung HS, Lee MY. Cancer screening through surface-enhanced Raman spectroscopy fingerprinting analysis of urinary metabolites using surface-carbonized silver nanowires on a filter membrane. Anal Chim Acta 2024; 1292:342233. [PMID: 38309850 DOI: 10.1016/j.aca.2024.342233] [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: 08/04/2023] [Revised: 11/05/2023] [Accepted: 01/09/2024] [Indexed: 02/05/2024]
Abstract
BACKGROUND Label-free surface-enhanced Raman spectroscopy (SERS)-based metabolic profiling has great potential for early cancer diagnosis, but further advancements in analytical methods and clinical evidence studies are required for clinical applications. To improve the cancer diagnostic accuracy of label-free SERS spectral analysis of complex biological fluids, it is necessary to obtain specifically enhanced SERS signals of cancer-related metabolites present at low concentrations. RESULTS This study presents a novel 3D SERS sensor, comprising a surface-carbonized silver nanowire (AgNW)-stacked filter membrane, alongside an optimized urine/methanol/chloroform extraction technique, which specifically changes the molecular adsorption and orientation of aromatic metabolites onto SERS substrates. By analyzing the pretreated urine samples on the surface-carbonized AgNW 3D SERS sensor, distinct and highly enhanced SERS peaks derived from semi-polar aromatic metabolites were observed for pancreatic cancer and prostate cancer samples compared with normal controls. Urine metabolite analysis using SERS fingerprinting successfully differentiated pancreatic cancer and prostate cancer groups from normal control group: normal control (n = 56), pancreatic cancer (n = 40), and prostate cancer (n = 39). SIGNIFICANCE AND NOVELTY We confirmed the clinical feasibility of performing fingerprint analysis of urinary metabolites based on the surface-carbonized AgNW 3D SERS sensor and methanol/chloroform extraction for noninvasive cancer screening. This technology holds potential for large-scale screening owing to its high accuracy, and cost effective, simple and rapid detection method.
Collapse
Affiliation(s)
- Ho-Jae Yu
- Medical Device Research Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Eunji Jang
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea
| | - Ayoung Woo
- Medical Device Research Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - In Woong Han
- Division of Hepato Biliary Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Hwang Gyun Jeon
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Vo Thi Nhat Linh
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea
| | - Sung-Gyu Park
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea
| | - Ho Sang Jung
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea.
| | - Min-Young Lee
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea.
| |
Collapse
|
19
|
Wang G, Lei W, Duan S, Cao A, Shi H. Preoperative evaluating early recurrence in resectable pancreatic ductal adenocarcinoma by using CT radiomics. Abdom Radiol (NY) 2024; 49:484-491. [PMID: 37955726 DOI: 10.1007/s00261-023-04074-x] [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: 07/10/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 11/14/2023]
Abstract
OBJECTIVE To investigate the feasibility of a radiomics model based on contrast-enhanced CT for preoperatively predicting early recurrence after curative resection in patients with resectable pancreatic ductal adenocarcinoma (PDAC). METHODS One hundred and eighty-six patients with resectable PDAC who underwent curative resection were included and allocated to training set (131 patients) and validation set (55 patients). Radiomics features were extracted from arterial phase and portal venous phase images. The Mann-Whitney U test and least absolute shrinkage and selection operator (LASSO) regression were used for feature selection and radiomics signature construction. The radiomics model based on radiomics signature and clinical features was developed by the multivariate logistic regression analysis. Performance of the radiomics model was investigated by the area under the receiver operating characteristic (ROC) curve. RESULTS The radiomics signature, consisting of three arterial phase and three venous phase features, showed optimal prediction performance for early recurrence in both training (AUC = 0.73) and validation sets (AUC = 0.66). Multivariate logistic analysis identified the radiomics signature (OR, 2.58; 95% CI 2.36-3.17; p = 0.002) and clinical stage (OR, 1.60; 95% CI 1.15-2.30; p = 0.007) as independent predictors. The AUC values for risk evaluation of early recurrence using the radiomics model incorporating clinical stage were 0.80 (training set) and 0.75 (validation set). CONCLUSION The radiomics-based model integrating with clinical stage can predict early recurrence after upfront surgery in patients with resectable PDAC.
Collapse
Affiliation(s)
- Gang Wang
- Department of Radiotherapy, The Second Affiliated Hospital of Xuzhou Medical University, 32 Meijian Road, Xuzhou, People's Republic of China
| | - Weijie Lei
- Department of Radiotherapy, The Second Affiliated Hospital of Xuzhou Medical University, 32 Meijian Road, Xuzhou, People's Republic of China
| | - Shaofeng Duan
- GE Healthcare, Pudong New Town, 1 Huatuo Road, Shanghai, People's Republic of China
| | - Aihong Cao
- Department of Radiology, The Second Affiliated Hospital of Xuzhou Medical University, 32 Meijian Road, Xuzhou, People's Republic of China.
| | - Hongyuan Shi
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, People's Republic of China.
| |
Collapse
|
20
|
Yan X, Fu X, Gui Y, Chen X, Cheng Y, Dai M, Wang W, Xiao M, Tan L, Zhang J, Shao Y, Wang H, Chang X, Lv K. Development and validation of a nomogram model based on pretreatment ultrasound and contrast-enhanced ultrasound to predict the efficacy of neoadjuvant chemotherapy in patients with borderline resectable or locally advanced pancreatic cancer. Cancer Imaging 2024; 24:13. [PMID: 38245789 PMCID: PMC10800053 DOI: 10.1186/s40644-024-00662-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 01/11/2024] [Indexed: 01/22/2024] Open
Abstract
OBJECTIVES To develop a nomogram using pretreatment ultrasound (US) and contrast-enhanced ultrasound (CEUS) to predict the clinical response of neoadjuvant chemotherapy (NAC) in patients with borderline resectable pancreatic cancer (BRPC) or locally advanced pancreatic cancer (LAPC). METHODS A total of 111 patients with pancreatic ductal adenocarcinoma (PDAC) treated with NAC between October 2017 and February 2022 were retrospectively enrolled. The patients were randomly divided (7:3) into training and validation cohorts. The pretreatment US and CEUS features were reviewed. Univariate and multivariate logistic regression analyses were used to determine the independent predictors of clinical response in the training cohort. Then a prediction nomogram model based on the independent predictors was constructed. The area under the curve (AUC), calibration plot, C-index and decision curve analysis (DCA) were used to assess the nomogram's performance, calibration, discrimination and clinical benefit. RESULTS The multivariate logistic regression analysis showed that the taller-than-wide shape in the longitudinal plane (odds ratio [OR]:0.20, p = 0.01), time from injection of contrast agent to peak enhancement (OR:3.64; p = 0.05) and Peaktumor/ Peaknormal (OR:1.51; p = 0.03) were independent predictors of clinical response to NAC. The predictive nomogram developed based on the above imaging features showed AUCs were 0.852 and 0.854 in the primary and validation cohorts, respectively. Good calibration was achieved in the training datasets, with C-index of 0.852. DCA verified the clinical usefulness of the nomogram. CONCLUSIONS The nomogram based on pretreatment US and CEUS can effectively predict the clinical response of NAC in patients with BRPC and LAPC; it may help guide personalized treatment.
Collapse
Affiliation(s)
- Xiaoyi Yan
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xianshui Fu
- Department of Ultrasound, No.304 Hospital of Chinese PLA, Beijing, 100037, China
| | - Yang Gui
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xueqi Chen
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yuejuan Cheng
- Department of Medical Oncology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Menghua Dai
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Weibin Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Mengsu Xiao
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Li Tan
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jing Zhang
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yuming Shao
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Huanyu Wang
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiaoyan Chang
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Ke Lv
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
21
|
McKay SC, Pathak S, Roberts KJ. Evaluation of post-operative surveillance strategies and surgeon perceptions and beliefs of surveillance for pancreatic ductal adenocarcinoma in the UK. HPB (Oxford) 2023; 25:1247-1254. [PMID: 37357113 DOI: 10.1016/j.hpb.2023.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/23/2023] [Accepted: 06/10/2023] [Indexed: 06/27/2023]
Abstract
BACKGROUND Despite high rates of recurrence after surgery for pancreatic ductal adenocarcinoma (PDAC) there is lack of standardised surveillance practices. We aimed to identify UK surveillance practice and interrogate surgeon beliefs around surveillance. METHODS A web-based survey was sent to all UK pancreatic units to assess surveillance practice for resected PDAC, factors influencing surveillance protocols, and perceptions and beliefs surrounding on current postoperative surveillance. RESULTS There was wide variation in reported practice between 40 consultant surgeons from 28 pancreatic units (100% unit response rate). 26% had standardised surveillance compared to 18% with no standardised practice. 16% individualised surveillance to the patient, and 40% reported differing practices between surgeons within units despite local surveillance protocols. 66% felt surveillance should be tailored to patient factors, and 58% to patient preference. There was a broad belief regarding a lack of robust evidence supporting surveillance making a trial necessary. Thematic analysis identified surveillance barriers, considerations for trial design, necessity for patient engagement and potential benefits of surveillance. DISCUSSION Wide variation in surveillance practice exists within and between units. A surveillance trial was deemed beneficial, however identified barriers potentially preclude a trial. Future work should assess acceptability for patients including impact on anxiety and quality-of-life.
Collapse
Affiliation(s)
- Siobhan C McKay
- Liver and Pancreas Unit, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham, UK; Department of Academic Surgery, University of Birmingham, UK
| | | | - Keith J Roberts
- Liver and Pancreas Unit, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham, UK.
| |
Collapse
|
22
|
Yugawa K, Maeda T, Nagata S, Sakai A, Taketani K, Yamaguchi S, Konishi K, Hashimoto K. A novel combined carbohydrate antigen 19-9 and lymphocyte-to-monocyte ratio score can predict early recurrence of resectable pancreatic ductal adenocarcinoma. Surg Today 2023; 53:1199-1208. [PMID: 36943449 DOI: 10.1007/s00595-023-02675-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/24/2023] [Indexed: 03/23/2023]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) often recurs early after radical resection, which causes a poor prognosis. This study aimed to establish a scoring model to assess the optimal treatment in patients who underwent surgery for PDAC. METHODS This single-center retrospective study included 127 patients who underwent radical resection for PDAC between 2005 and 2021. Early recurrence (ER) was defined as recurrence within 12 months after resection. The predictive effect for ER was evaluated using receiver operating characteristic (ROC) curves of preoperative parameters. RESULTS ER occurred in 43 (33.9%) patients. The ER group had a significantly worse prognosis than the non-ER group (p < 0.0001). The carbohydrate antigen 19-9 (CA19-9) level and lymphocyte-to-monocyte ratio (LMR) were the strongest diagnostic factors (areas under the ROC curves: 0.74 and 0.68, respectively). The ER prediction score was calculated using optimal cutoff values. A higher CA19-9-LMR score was associated with a worse prognosis in terms of the overall and recurrence-free survival (p = 0.0017 and p < 0.0001, respectively). A multivariate analysis identified a high CA19-9-LMR score as an independent predictor of ER. CONCLUSIONS The CA19-9-LMR scoring model can predict ER after surgery and is applicable for risk stratification in the assessment of patients with resectable PDAC.
Collapse
Affiliation(s)
- Kyohei Yugawa
- Department of Surgery, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Hiroshima, 730-0052, Japan
| | - Takashi Maeda
- Department of Surgery, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Hiroshima, 730-0052, Japan.
| | - Shigeyuki Nagata
- Department of Surgery, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Hiroshima, 730-0052, Japan
| | - Akihiro Sakai
- Department of Surgery, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Hiroshima, 730-0052, Japan
| | - Kenji Taketani
- Department of Surgery, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Hiroshima, 730-0052, Japan
| | - Shohei Yamaguchi
- Department of Surgery, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Hiroshima, 730-0052, Japan
| | - Kozo Konishi
- Department of Surgery, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Hiroshima, 730-0052, Japan
| | - Kenkichi Hashimoto
- Department of Surgery, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Hiroshima, 730-0052, Japan
| |
Collapse
|
23
|
Dao J, Conway PJ, Subramani B, Meyyappan D, Russell S, Mahadevan D. Using cfDNA and ctDNA as Oncologic Markers: A Path to Clinical Validation. Int J Mol Sci 2023; 24:13219. [PMID: 37686024 PMCID: PMC10487653 DOI: 10.3390/ijms241713219] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The detection of circulating tumor DNA (ctDNA) in liquid biopsy samples as an oncological marker is being used in clinical trials at every step of clinical management. As ctDNA-based liquid biopsy kits are developed and used in clinics, companies work towards increased convenience, accuracy, and cost over solid biopsies and other oncological markers. The technology used to differentiate ctDNA and cell-free DNA (cfDNA) continues to improve with new tests and methodologies being able to detect down to mutant allele frequencies of 0.001% or 1/100,000 copies. Recognizing this development in technology, the FDA has recently given pre-market approval and breakthrough device designations to multiple companies. The purpose of this review is to look at the utility of measuring total cfDNA, techniques used to differentiate ctDNA from cfDNA, and the utility of different ctDNA-based liquid biopsy kits using relevant articles from PubMed, clinicaltrials.gov, FDA approvals, and company newsletters. Measuring total cfDNA could be a cost-effective, viable prognostic marker, but various factors do not favor it as a monitoring tool during chemotherapy. While there may be a place in the clinic for measuring total cfDNA in the future, the lack of standardization means that it is difficult to move forward with large-scale clinical validation studies currently. While the detection of ctDNA has promising standardized liquid biopsy kits from various companies with large clinical trials ongoing, their applications in screening and minimal residual disease can suffer from lower sensitivity. However, researchers are working towards solutions to these issues with innovations in technology, multi-omics, and sampling. With great promise, further research is needed before liquid biopsies can be recommended for everyday clinical management.
Collapse
Affiliation(s)
- Jonathan Dao
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Patrick J. Conway
- Mays Cancer Center, University of Texas Health, San Antonio, TX 78229, USA
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Baskaran Subramani
- Mays Cancer Center, University of Texas Health, San Antonio, TX 78229, USA
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Devi Meyyappan
- Mays Cancer Center, University of Texas Health, San Antonio, TX 78229, USA
| | - Sammy Russell
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Daruka Mahadevan
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
- Mays Cancer Center, University of Texas Health, San Antonio, TX 78229, USA
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
24
|
Linscott MP, Markus H, Sennett M, Abendroth C, Yee NS. Nab-Paclitaxel and Gemcitabine as First-Line Treatment of Metastatic Ampullary Adenocarcinoma with a Novel R-Spondin2 RNA Fusion and NTRK3 Mutation. Biomedicines 2023; 11:2326. [PMID: 37626821 PMCID: PMC10452745 DOI: 10.3390/biomedicines11082326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Ampullary adenocarcinoma is a rare malignancy that lacks standard systemic treatment. We describe a case of recurrent metastatic ampullary adenocarcinoma of the pancreaticobiliary subtype treated with nanoparticle albumin-bound (nab)-paclitaxel and gemcitabine as first-line treatment. This report also highlights the molecular profile of the ampullary adenocarcinoma and circulating tumor DNA (ctDNA). This is a case of pancreaticobiliary ampullary adenocarcinoma in a 67-year-old woman who initially presented with painless jaundice. Endoscopic and imaging evaluation revealed biliary ductal dilation secondary to an ampullary mass. Pathology confirmed the diagnosis of ampullary adenocarcinoma of the pancreaticobiliary subtype. She underwent surgical resection of the tumor, followed by adjuvant chemotherapy with gemcitabine and capecitabine. The tumor subsequently recurred in the liver. She received palliative chemotherapy with nab-paclitaxel and gemcitabine, resulting in an objective tumor response for 14 months. Molecular profiling of the tumor and ctDNA revealed a novel MATN2-RSPO RNA fusion and a novel NTRK3 mutation, respectively. Our report suggests that long-term durable response can be achieved in metastatic pancreaticobiliary ampullary adenocarcinoma using nab-paclitaxel and gemcitabine. Molecular profiling of the tumor identified a novel R-Spondin2 RNA fusion and NTRK3 mutation that can be potentially targeted for treatment.
Collapse
Affiliation(s)
- Maryknoll P. Linscott
- Medical Scientist Training Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.P.L.); (H.M.); (M.S.)
| | - Havell Markus
- Medical Scientist Training Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.P.L.); (H.M.); (M.S.)
| | - Mackenzie Sennett
- Medical Scientist Training Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.P.L.); (H.M.); (M.S.)
| | - Catherine Abendroth
- Department of Pathology, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA;
| | - Nelson S. Yee
- Division of Hematology-Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Next-Generation Therapies Program, Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| |
Collapse
|
25
|
Schepis T, De Lucia SS, Pellegrino A, Del Gaudio A, Maresca R, Coppola G, Chiappetta MF, Gasbarrini A, Franceschi F, Candelli M, Nista EC. State-of-the-Art and Upcoming Innovations in Pancreatic Cancer Care: A Step Forward to Precision Medicine. Cancers (Basel) 2023; 15:3423. [PMID: 37444534 DOI: 10.3390/cancers15133423] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Pancreatic cancer remains a social and medical burden despite the tremendous advances that medicine has made in the last two decades. The incidence of pancreatic cancer is increasing, and it continues to be associated with high mortality and morbidity rates. The difficulty of early diagnosis (the lack of specific symptoms and biomarkers at early stages), the aggressiveness of the disease, and its resistance to systemic therapies are the main factors for the poor prognosis of pancreatic cancer. The only curative treatment for pancreatic cancer is surgery, but the vast majority of patients with pancreatic cancer have advanced disease at the time of diagnosis. Pancreatic surgery is among the most challenging surgical procedures, but recent improvements in surgical techniques, careful patient selection, and the availability of minimally invasive techniques (e.g., robotic surgery) have dramatically reduced the morbidity and mortality associated with pancreatic surgery. Patients who are not candidates for surgery may benefit from locoregional and systemic therapy. In some cases (e.g., patients for whom marginal resection is feasible), systemic therapy may be considered a bridge to surgery to allow downstaging of the cancer; in other cases (e.g., metastatic disease), systemic therapy is considered the standard approach with the goal of prolonging patient survival. The complexity of patients with pancreatic cancer requires a personalized and multidisciplinary approach to choose the best treatment for each clinical situation. The aim of this article is to provide a literature review of the available treatments for the different stages of pancreatic cancer.
Collapse
Affiliation(s)
- Tommaso Schepis
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Sara Sofia De Lucia
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Antonio Pellegrino
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Angelo Del Gaudio
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Rossella Maresca
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Gaetano Coppola
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Michele Francesco Chiappetta
- Section of Gastroenterology and Hepatology, Promise, Policlinico Universitario Paolo Giaccone, 90127 Palermo, Italy
- IBD-Unit, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Antonio Gasbarrini
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Anesthesiological and Reanimation Sciences, Fondazione Universitaria Policlinico Agostino Gemelli di Roma, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Marcello Candelli
- Department of Emergency Anesthesiological and Reanimation Sciences, Fondazione Universitaria Policlinico Agostino Gemelli di Roma, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Enrico Celestino Nista
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| |
Collapse
|
26
|
Imran KM, Tintera B, Morrison HA, Tupik JD, Nagai-Singer MA, Ivester H, Council-Troche M, Edwards M, Coutermarsh-Ott S, Byron C, Clark-Deener S, Uh K, Lee K, Boulos P, Rowe C, Coviello C, Allen IC. Improved Therapeutic Delivery Targeting Clinically Relevant Orthotopic Human Pancreatic Tumors Engrafted in Immunocompromised Pigs Using Ultrasound-Induced Cavitation: A Pilot Study. Pharmaceutics 2023; 15:1585. [PMID: 37376034 PMCID: PMC10302458 DOI: 10.3390/pharmaceutics15061585] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/03/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Pancreatic tumors can be resistant to drug penetration due to high interstitial fluid pressure, dense stroma, and disarrayed vasculature. Ultrasound-induced cavitation is an emerging technology that may overcome many of these limitations. Low-intensity ultrasound, coupled with co-administered cavitation nuclei consisting of gas-stabilizing sub-micron scale SonoTran Particles, is effective at increasing therapeutic antibody delivery to xenograft flank tumors in mouse models. Here, we sought to evaluate the effectiveness of this approach in situ using a large animal model that mimics human pancreatic cancer patients. Immunocompromised pigs were surgically engrafted with human Panc-1 pancreatic ductal adenocarcinoma (PDAC) tumors in targeted regions of the pancreas. These tumors were found to recapitulate many features of human PDAC tumors. Animals were intravenously injected with the common cancer therapeutics Cetuximab, gemcitabine, and paclitaxel, followed by infusion with SonoTran Particles. Select tumors in each animal were targeted with focused ultrasound to induce cavitation. Cavitation increased the intra-tumor concentrations of Cetuximab, gemcitabine, and paclitaxel by 477%, 148%, and 193%, respectively, compared to tumors that were not targeted with ultrasound in the same animals. Together, these data show that ultrasound-mediated cavitation, when delivered in combination with gas-entrapping particles, improves therapeutic delivery in pancreatic tumors under clinically relevant conditions.
Collapse
Affiliation(s)
- Khan Mohammad Imran
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA 24061, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Benjamin Tintera
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Holly A. Morrison
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Juselyn D. Tupik
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Margaret A. Nagai-Singer
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Hannah Ivester
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA 24061, USA
| | - McAlister Council-Troche
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Michael Edwards
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Sheryl Coutermarsh-Ott
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Christopher Byron
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Sherrie Clark-Deener
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Kyungjun Uh
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO 65211, USA
| | - Kiho Lee
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO 65211, USA
| | - Paul Boulos
- OxSonics Therapeutics, Oxford Science Park, Oxford OX4 4GA, UK
| | - Cliff Rowe
- OxSonics Therapeutics, Oxford Science Park, Oxford OX4 4GA, UK
| | | | - Irving C. Allen
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA 24061, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| |
Collapse
|
27
|
Botta GP, Huynh TR, Spierling‐Bagsic SR, Agelidis A, Schaffer R, Lin R, Sigal D. Neoadjuvant chemotherapy and radiotherapy outcomes in borderline-resectable and locally-advanced pancreatic cancer patients. Cancer Med 2023; 12:7713-7723. [PMID: 36478411 PMCID: PMC10134275 DOI: 10.1002/cam4.5523] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 11/07/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND There is no agreed upon standard of care for borderline-resectable pancreatic cancer (BRPC) or locally-advanced pancreatic cancer (LAPC) patients regarding the benefit of chemotherapy or radiation alone or in combination. PATIENTS AND METHODS We completed a retrospective cohort analysis of BRPC and LAPC patients at a cancer center with expertise in multi-disciplinary pancreatic ductal adenocarcinoma (PDAC) treatment over a 5-year period from 03/01/2014 to 03/01/2019 (cut-off date). The total evaluable newly diagnosed, treatment naïve, BRPC, and LAPC patients with adequate organ function and ability to obtain treatment after multidisciplinary review was 52 patients. After analysis, patients were evaluated for rates of resection, extent of resection (R0 or R1), median progression-free survival (mPFS), and median overall survival (mOS). RESULTS Patients were treated with chemotherapy alone (gemcitabine and nab-paclitaxel = 77% (20/26); FOLFIRINOX = 19% (5/26); single agent gemcitabine 3.8% (1/26)), or chemotherapy followed by chemoradiation (gemcitabine +5 Gy × 5 weeks), or chemoradiation alone prior to re-staging and potential resection. Of the 29% (15/52) of patients who went on to surgical resection, 73% (11/15) achieved R0 resection. An R0 resection was achieved in 35% (9/26) of patients treated with chemotherapy alone, 7.6% (1/13) in a patient treated with chemotherapy followed by radiation, and 7.6% (1/13) with concurrent chemoradiotherapy alone. Chemotherapy alone achieved a mPFS of 16.4 months (p < 0.0025) and mOS of 26.2 months (p < 0.0001), chemotherapy followed by chemoradiation was 13.0 months and 14.9 months respectively, while concurrent chemoradiotherapy was 6.9 months and 7.3 months. CONCLUSIONS AND RELEVANCE BRPC and LAPC patients capable of surgery after only receiving neoadjuvant treatment with chemotherapy had higher rates of R0 resection with prolonged median PFS and OS compared with any patient needing combination chemotherapy with radiotherapy.
Collapse
Affiliation(s)
- Gregory P. Botta
- Division of Hematology/Oncology, Department of Medicine, Moores Cancer CenterUniversity of California San DiegoLa JollaCaliforniaUSA
- Division of Medical OncologyScripps MD Anderson Cancer CenterLa JollaCaliforniaUSA
- Scripps Research Translational InstituteLa JollaCaliforniaUSA
| | - Tridu R. Huynh
- Division of Hematology/Oncology, Department of Medicine, Moores Cancer CenterUniversity of California San DiegoLa JollaCaliforniaUSA
- Scripps Research Translational InstituteLa JollaCaliforniaUSA
- Division of Internal MedicineScripps Clinic/Green HospitalLa JollaCaliforniaUSA
| | | | - Alexander Agelidis
- Scripps Research Translational InstituteLa JollaCaliforniaUSA
- Division of Internal MedicineScripps Clinic/Green HospitalLa JollaCaliforniaUSA
| | - Randolph Schaffer
- Division of Hepatopancreatobiliary SurgeryScripps MD Anderson Cancer CenterLa JollaCaliforniaUSA
| | - Ray Lin
- Division of Radiation OncologyScripps MD Anderson Cancer CenterLa JollaCaliforniaUSA
| | - Darren Sigal
- Division of Medical OncologyScripps MD Anderson Cancer CenterLa JollaCaliforniaUSA
| |
Collapse
|
28
|
Kung H, Yu J. Targeted therapy for pancreatic ductal adenocarcinoma: Mechanisms and clinical study. MedComm (Beijing) 2023; 4:e216. [PMID: 36814688 PMCID: PMC9939368 DOI: 10.1002/mco2.216] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 02/21/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive and lethal malignancy with a high rate of recurrence and a dismal 5-year survival rate. Contributing to the poor prognosis of PDAC is the lack of early detection, a complex network of signaling pathways and molecular mechanisms, a dense and desmoplastic stroma, and an immunosuppressive tumor microenvironment. A recent shift toward a neoadjuvant approach to treating PDAC has been sparked by the numerous benefits neoadjuvant therapy (NAT) has to offer compared with upfront surgery. However, certain aspects of NAT against PDAC, including the optimal regimen, the use of radiotherapy, and the selection of patients that would benefit from NAT, have yet to be fully elucidated. This review describes the major signaling pathways and molecular mechanisms involved in PDAC initiation and progression in addition to the immunosuppressive tumor microenvironment of PDAC. We then review current guidelines, ongoing research, and future research directions on the use of NAT based on randomized clinical trials and other studies. Finally, the current use of and research regarding targeted therapy for PDAC are examined. This review bridges the molecular understanding of PDAC with its clinical significance, development of novel therapies, and shifting directions in treatment paradigm.
Collapse
Affiliation(s)
- Heng‐Chung Kung
- Krieger School of Arts and SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jun Yu
- Departments of Medicine and OncologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
29
|
Design and Assessment of a Novel Biconical Human-Sized Alternating Magnetic Field Coil for MNP Hyperthermia Treatment of Deep-Seated Cancer. Cancers (Basel) 2023; 15:cancers15061672. [PMID: 36980560 PMCID: PMC10046348 DOI: 10.3390/cancers15061672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/07/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
Magnetic nanoparticle (MNP) hyperthermia therapy is a treatment technique that can be used alone or as an adjunct to radiation and/or chemotherapies for killing cancer cells. During treatment, MNPs absorb a part of electromagnetic field (EMF) energy and generate localized heat when subjected to an alternating magnetic field (AMF). The MNP-absorbed EMF energy, which is characterized by a specific absorption rate (SAR), is directly proportional to AMF frequency and the magnitude of transmitting currents in the coil. Furthermore, the AMF penetrates inside tissue and induces eddy currents in electrically conducting tissues, which are proportional to the electric field (J = σE). The eddy currents produce Joule heating (<J·E> = 0.5·σ·E2) in the normal tissue, the rate of energy transfer to the charge carriers from the applied electric fields. This Joule heating contains only the electric field because the magnetic field is always perpendicular to the velocity of the conduction charges, i.e., it does not produce work on moving charge. Like the SAR due to MNP, the electric field produced by the AMF coil is directly proportional to AMF frequency and the magnitude of transmitting currents in the coil. As a result, the Joule heating is directly proportional to the square of the frequency and transmitter current magnitude. Due to the fast decay of magnetic fields from an AMF coil over distance, MNP hyperthermia treatment of deep-seated tumors requires high-magnitude transmitting currents in the coil for clinically achievable MNP distributions in the tumor. This inevitably produces significant Joule heating in the normal tissue and becomes more complicated for a standard MNP hyperthermia approach for deep-seated tumors, such as pancreatic, prostate, liver, lung, ovarian, kidney, and colorectal cancers. This paper presents a novel human-sized AMF coil and MNP hyperthermia system design for safely and effectively treating deep-seated cancers. The proposed design utilizes the spatial distribution of electric and magnetic fields of circular coils. Namely, it first minimizes the SAR due to eddy currents in the normal tissue by moving the conductors away from the tissue (i.e., increasing coils’ radii), and second, it increases the magnetic field at the targeted area (z = 0) due to elevated coils (|z| > 0) by increasing the radius of the elevated coils (|z| > 0). This approach is a promising alternative aimed at overcoming the limitation of standard MNP hyperthermia for deep-seated cancers by taking advantage of the transmitter coil’s electric and magnetic field distributions in the human body for maximizing AMF in tumor regions and avoiding damage to normal tissue. The human-sized coil’s AMF, MNP activation, and eddy current distribution characteristics are investigated for safe and effective treatment of deep-seated tumors using numerical models. Namely, computational results such as AMF, Joule heating SAR, and temperature distributions are presented for a full-body, 3D human model. The SAR and temperature distributions clearly show that the proposed human-sized AMF coil can provide clinically relevant AMF to the region occupied by deep-seated cancers for the application of MNP hyperthermia therapy while causing less Joule heating in the normal tissues than commonly used AMF techniques.
Collapse
|
30
|
Are Aspects of Integrative Concepts Helpful to Improve Pancreatic Cancer Therapy? Cancers (Basel) 2023; 15:cancers15041116. [PMID: 36831465 PMCID: PMC9953994 DOI: 10.3390/cancers15041116] [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: 12/29/2022] [Revised: 01/24/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Numerous clinical studies have been conducted to improve the outcomes of patients suffering from pancreatic cancer. Different approaches using targeted therapeutic strategies and precision medicine methods have been investigated, and synergies and further therapeutic advances may be achieved through combinations with integrative methods. For pancreatic tumors, a particular challenge is the presence of a microenvironment and a dense stroma, which is both a physical barrier to drug penetration and a complex entity being controlled by the immune system. Therefore, the state of immunological tolerance in the tumor microenvironment must be overcome, which is a considerable challenge. Integrative approaches, such as hyperthermia, percutaneous irreversible electroporation, intra-tumoral injections, phytotherapeutics, or vitamins, in combination with standard-oncological therapies, may potentially contribute to the control of pancreatic cancer. The combined application of standard-oncological and integrative methods is currently being studied in ongoing clinical trials. An actual overview is given here.
Collapse
|
31
|
Roberts DA, Watson E, Macdonald C, Khan Y, Prideaux S, Puthiyakunnel Saji A, Postaleniec E, Selvakumar J, Haghighat Ghahfarokhi M, Davidson B, Gurusamy K. Management of pain and cachexia in pancreatic cancer: Protocol for two systematic reviews, network meta-analysis, surveys and focus groups (Preprint). JMIR Res Protoc 2023; 12:e46335. [PMID: 37014692 PMCID: PMC10139686 DOI: 10.2196/46335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/26/2023] [Accepted: 02/26/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Approximately 75% of people with pancreatic cancer experience pain, and >50% of them have cachexia (weakness and wasting of the body). However, there is considerable uncertainty regarding the management of these distressing symptoms. OBJECTIVE Our primary objectives are to compare the relative benefits and harms of different interventions for pain in people with unresectable pancreatic cancer and for prevention and treatment of cachexia due to pancreatic cancer, through systematic reviews and network meta-analysis. Our secondary objectives are to develop an evidence-based clinical care pathway to manage pain and prevent and treat cachexia in people with pancreatic cancer through surveys and focus groups involving patients, carers, and health care professionals. METHODS We will perform 2 systematic reviews of the literature related to pain and cachexia in people with pancreatic cancer using searches from Cochrane Library, MEDLINE, Embase, Science Citation Index, and trial registries. Two researchers will independently screen for eligibility and identify randomized controlled trials (no language or publication status restriction), comparing interventions for pain or cachexia based on full-texts for articles shortlisted during screening. We will assess risk of bias in the trials using the Cochrane risk of bias tool (version 2.0) and obtain data related to baseline prognostic characteristics, potential effect modifiers and outcome data related to overall survival, health-related quality of life, treatment-related complications, and resource utilisation. We aim to conduct network meta-analysis on outcomes with multiple treatment comparisons where possible, otherwise, meta-analysis with direct comparisons, or narrative synthesis. We will perform various subgroup and sensitivity analyses. Using information obtained from both systematic reviews, we will conduct 2 surveys: one directed to patients or carers to assess acceptability of interventions, and the other to health care professionals to assess feasibility of delivery in the National Health Service. Four mixed focus groups will be conducted to evaluate findings and foster consensus in the development of the care pathway. RESULTS Funding was awarded from April 2022 (NIHR202727). Both systematic review protocols were prospectively registered on PROSPERO in May 2022. Formal searches began thereafter. Approval by the University College London Research Ethics Committee (23563/001) was received in December 2022. Data collection began in January 2023; data analysis will begin in May 2023 (completion expected by October 2023). CONCLUSIONS This study will comprehensively encompass major interventions for management of pain in people with unresectable pancreatic cancer, and prevention and treatment of cachexia in people with pancreatic cancer. Key stakeholders will facilitate the development of an evidence-based care pathway, ensuring both acceptability and feasibility. The project ends in April 2024 and published results are expected within 12 months of completion. We aim to present the findings through patient group websites, conferences, and publications, irrespective of the findings, in a peer-reviewed journal. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/46335.
Collapse
Affiliation(s)
- Danielle Amanda Roberts
- Division of Surgery and Interventional Science, Hampstead Campus, University College London, London, United Kingdom
| | - Eila Watson
- Supportive Cancer Care Research Group, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, United Kingdom
| | | | | | - Sarah Prideaux
- Patient and Public Involvement Representative, England, United Kingdom
| | | | - Emilia Postaleniec
- Leicester Medical School, The University of Leicester, Leicester, United Kingdom
| | - Jashan Selvakumar
- St George's, University of London Medical School, London, United Kingdom
| | | | - Brian Davidson
- Division of Surgery and Interventional Science, Hampstead Campus, University College London, London, United Kingdom
| | - Kurinchi Gurusamy
- Division of Surgery and Interventional Science, Hampstead Campus, University College London, London, United Kingdom
| |
Collapse
|
32
|
Søreide K, Rangelova E, Dopazo C, Mieog S, Stättner S. Pancreatic cancer. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2023; 49:521-525. [PMID: 36604234 DOI: 10.1016/j.ejso.2023.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/01/2023] [Indexed: 01/04/2023]
Abstract
The need for a common education and training track in surgical oncology across Europe has been emphasized. ESSO provides several hands-on courses for skills training and face-to-face discussions. The core curriculum provides a framework for the overall theoretical requirements in surgical oncology. The UEMS/EBSQ fellowship exam is designed to test core competencies in the candidate's core knowledge in their prespecified area of expertise. A core set of points for each cancer type is lacking. Hence, a condensed outline of themed expected to be covered in the curriculum and relevant to an optimal practice in surgical oncology is provided. This article outlines pancreatic cancer.
Collapse
Affiliation(s)
- Kjetil Søreide
- Department of Gastrointestinal Surgery, HPB Unit, Stavanger University Hospital, Stavanger, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - Elena Rangelova
- Section of Upper GI Surgery at Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Christina Dopazo
- Department of HPB Surgery and Transplants, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Sven Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Stefan Stättner
- Department of General, Visceral and Vascular Surgery, Salzkammergut Klinikum, OÖG, Dr. Wilhelm Bock Strasse 1, 4840, Vöcklabruck, Austria
| |
Collapse
|
33
|
Yang P, Mao K, Gao Y, Wang Z, Wang J, Chen Y, Ma C, Bian Y, Shao C, Lu J. Tumor size measurements of pancreatic cancer with neoadjuvant therapy based on RECIST guidelines: is MRI as effective as CT? Cancer Imaging 2023; 23:8. [PMID: 36653861 PMCID: PMC9850516 DOI: 10.1186/s40644-023-00528-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVES To compare tumor size measurements using CT and MRI in pancreatic cancer (PC) patients with neoadjuvant therapy (NAT). METHODS This study included 125 histologically confirmed PC patients who underwent NAT. The tumor sizes from CT and MRI before and after NAT were compared by using Bland-Altman analyses and intraclass correlation coefficients (ICCs). Variations in tumor size estimates between MRI and CT in relationship to different factors, including NAT methods (chemotherapy, chemoradiotherapy), tumor locations (head/neck, body/tail), tumor regression grade (TRG) levels (0-2, 3), N stages (N0, N1/N2) and tumor resection margin status (R0, R1), were further analysed. The McNemar test was used to compare the efficacy of NAT evaluations based on the CT and MRI measurements according to RECIST 1.1 criteria. RESULTS There was no significant difference between the median tumor sizes from CT and MRI before and after NAT (P = 0.44 and 0.39, respectively). There was excellent agreement in tumor size between MRI and CT, with mean size differences and limits of agreement (LOAs) of 1.5 [-9.6 to 12.7] mm and 0.9 [-12.6 to 14.5] mm before NAT (ICC, 0.93) and after NAT (ICC, 0.91), respectively. For all the investigated factors, there was good or excellent correlation (ICC, 0.76 to 0.95) for tumor sizes between CT and MRI. There was no significant difference in the efficacy evaluation of NAT between CT and MRI measurements (P = 1.0). CONCLUSION MRI and CT have similar performance in assessing PC tumor size before and after NAT.
Collapse
Affiliation(s)
- Panpan Yang
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Kuanzheng Mao
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China ,grid.267139.80000 0000 9188 055XSchool of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yisha Gao
- grid.73113.370000 0004 0369 1660Department of Pathology, Changhai Hospital of Shanghai, Naval Medical University, Shanghai, China
| | - Zhen Wang
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Jun Wang
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Yufei Chen
- grid.24516.340000000123704535College of Electronic and Information Engineering, Tongji University, Shanghai, China
| | - Chao Ma
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China ,grid.24516.340000000123704535College of Electronic and Information Engineering, Tongji University, Shanghai, China
| | - Yun Bian
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Chengwei Shao
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Jianping Lu
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| |
Collapse
|
34
|
Adam MA, Glencer A, AlMasri S, Winters S, Bahary N, Singhi A, Lee KK, Paniccia A, Zureikat AH. Neoadjuvant Therapy Versus Upfront Resection for Nonpancreatic Periampullary Adenocarcinoma. Ann Surg Oncol 2023; 30:165-174. [PMID: 35925536 PMCID: PMC11186695 DOI: 10.1245/s10434-022-12257-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/30/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND In contrast to pancreatic ductal adenocarcinoma (PDAC), neoadjuvant therapy (NAT) for periampullary adenocarcinomas is not well studied, with data limited to single-institution retrospective reviews with small cohorts. We sought to compare outcomes of NAT versus upfront resection (UR) for non-PDAC periampullary adenocarcinomas. PATIENTS AND METHODS Using the National Cancer Database (NCDB), we identified patients who underwent surgery for extrahepatic cholangiocarcinoma, ampullary adenocarcinoma, or duodenal adenocarcinoma from 2006 to 2016. We compared outcomes between NAT versus UR groups for each tumor subtype with 1:3 propensity score matching. Cox regression was used to identify predictors of survival. RESULTS Among 7656 patients who underwent resection for non-PDAC periampullary adenocarcinoma, the proportion of patients who received NAT increased from 6 to 11% for cholangiocarcinoma (p < 0.01), 1 to 4% for ampullary adenocarcinoma (p = 0.01), and 5 to 8% for duodenal adenocarcinoma (p = 0.08). Length of stay, readmission, and 30-day mortality were comparable between NAT and UR. All tumor subtypes were downstaged following NAT (p < 0.01). The R0 resection rate was significantly higher in patients with extrahepatic cholangiocarcinoma who received NAT, and these patients had improved median overall survival (38 vs 26 months, p < 0.001). After adjustment for clinicopathologic factors and adjuvant chemotherapy, use of NAT was associated with improved survival in patients with cholangiocarcinoma [hazard ratio (HR) 0.69, 95% confidence interval (CI) 0.54-0.89, p = 0.004] but not duodenal or ampullary adenocarcinoma. The survival advantage for cholangiocarcinoma persisted after propensity matching. CONCLUSION This national cohort analysis suggests, for the first time, that neoadjuvant therapy is associated with improved survival in patients with extrahepatic cholangiocarcinoma.
Collapse
Affiliation(s)
- Mohamed Abdelgadir Adam
- Division of Surgical Oncology, University of California, San Francisco, San Francisco, CA, USA.
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA.
| | - Alexa Glencer
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Samer AlMasri
- UPMC Network Cancer Registry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sharon Winters
- UPMC Network Cancer Registry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nathan Bahary
- Department of Internal Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aatur Singhi
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kenneth K Lee
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Amer H Zureikat
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
35
|
Sardar M, Recio-Boiles A, Mody K, Karime C, Chandana SR, Mahadevan D, Starr J, Jones J, Borad M, Babiker H. Pharmacotherapeutic options for pancreatic ductal adenocarcinoma. Expert Opin Pharmacother 2022; 23:2079-2089. [PMID: 36394449 DOI: 10.1080/14656566.2022.2149322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy projected to be the 2nd leading cause of cancer related death in the USA by 2030. This manuscript discusses current and evolving treatment approaches in patients with pancreatic cancer. AREAS COVERED PDAC is classified as: a) resectable, b) borderline resectable, c) unresectable (locally advanced and metastatic). The standard of care for patients who present with resectable pancreatic adenocarcinoma is six months of adjuvant modified (m) FOLFIRINOX, gemcitabine plus capecitabine, or single agent gemcitabine. For many reasons, there has been a paradigm shift to employing neoadjuvant chemotherapy. For resectable and borderline resectable patients, we generally start with systemic therapy and reevaluate resectability with subsequent scans specifically when the tumor is located in the head or body of the pancreas. Combined chemoradiation therapy can be employed in select patients. The standard of care for metastatic PDAC is FOLFIRINOX or gemcitabine and nab-paclitaxel. Germline and somatic genomic profiling should be obtained in all patients. Patients with a germline BRCA mutation can receive upfront gemcitabine and cisplatin. EXPERT OPINION Thorough understanding of molecular pathogenesis in PDAC has opened various therapeutic avenues. We remain optimistic that future treatment modalities such as targeted therapies, cellular therapies and immunotherapy will further improve survival in PDAC.
Collapse
Affiliation(s)
- Muhammad Sardar
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, Az, USA
| | - Alejandro Recio-Boiles
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, Az, USA
| | - Kabir Mody
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Cancer Center, Jacksonville, FL, USA
| | | | | | - Daruka Mahadevan
- Division of Hematology and Oncology, Department of Medicine, University of Texas, San Antonio, Texas, USA
| | - Jason Starr
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Cancer Center, Jacksonville, FL, USA
| | - Jeremy Jones
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Cancer Center, Jacksonville, FL, USA
| | - Mitesh Borad
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Cancer Center, Phoenix, AZ, USA
| | - Hani Babiker
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Cancer Center, Jacksonville, FL, USA
| |
Collapse
|
36
|
Kim H, Kim DH, Song IH, Kim B, Oh SN, Choi JI, Rha SE. Survival Prediction after Curative Resection of Pancreatic Ductal Adenocarcinoma by Imaging-Based Intratumoral Necrosis. Cancers (Basel) 2022; 14:cancers14225671. [PMID: 36428764 PMCID: PMC9688323 DOI: 10.3390/cancers14225671] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
We aimed to determine the histopathological characteristics and prognosis of curatively resected pancreatic ductal adenocarcinoma (PDAC) showing intratumoral necrosis on preoperative CT or MRI. This study consecutively included 102 patients who underwent upfront surgery with margin-negative resection from 2012 to 2020. All patients underwent both pancreatic CT and MRI within 1 month before surgery. Two radiologists independently assessed CT/MRI findings, including the presence of CT- and MRI-detected necrosis. Histopathological characteristics of PDACs according to CT or MRI detection of necrosis were evaluated. Disease-free survival (DFS) and overall survival (OS) were assessed by the Kaplan−Meier method and the Cox proportional hazards model. Among the 102 PDAC patients, 14 patients (13.7%) had CT-detected necrosis, and 16 patients (15.7%) had MRI-detected necrosis, of which 9 showed both CT- and MRI-detected necrosis. PDACs with CT- or MRI-detected necrosis demonstrated a significantly higher degree of histopathological necrosis than those without (p < 0.001). Multivariable analysis revealed that tumor size (hazard ratio [HR], 1.19; p = 0.040), tumor location (HR, 0.46; p = 0.009), and MRI-detected necrosis (HR, 2.64; p = 0.002) had independent associations with DFS. Only MRI-detected necrosis was significantly associated with OS (HR, 2.59; p = 0.004). Therefore, MRI-detected necrosis might be a potential imaging predictor of poor survival after curative resection of PDAC.
Collapse
Affiliation(s)
- Hokun Kim
- Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Dong Hwan Kim
- Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
- Correspondence: ; Tel.: +82-2-2258-1427; Fax: +82-2-599-6771
| | - In Hye Song
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-Ro 43-Gil, Songpa-Gu, Seoul 05505, Republic of Korea
| | - Bohyun Kim
- Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Soon Nam Oh
- Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Joon-Il Choi
- Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Sung Eun Rha
- Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| |
Collapse
|
37
|
Bertoni M, Bertoni C, Abatangelo S, Scatizzi M, Lotti P. A Case Report of New-Onset Diabetes Mellitus as an Early Warning Sign of Pancreatic Ductal Adenocarcinoma in an Elderly Patient: The Earlier the Diagnosis and Surgery, the Better the Prognosis. Cureus 2022; 14:e31608. [DOI: 10.7759/cureus.31608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2022] [Indexed: 11/18/2022] Open
|
38
|
Seufferlein T, Mayerle J, Böck S, Brunner T, Ettrich TJ, Grenacher L, Gress TM, Hackert T, Heinemann V, Kestler A, Sinn M, Tannapfel A, Wedding U, Uhl W. S3-Leitlinie zum exokrinen Pankreaskarzinom – Langversion 2.0 – Dezember 2021 – AWMF-Registernummer: 032/010OL. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2022; 60:e812-e909. [PMID: 36368658 DOI: 10.1055/a-1856-7346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | | | - Stefan Böck
- Medizinische Klinik und Poliklinik III, Universitätsklinikum München, Germany
| | - Thomas Brunner
- Universitätsklinik für Strahlentherapie-Radioonkologie, Medizinische Universität Graz, Austria
| | | | | | - Thomas Mathias Gress
- Klinik für Gastroenterologie und Endokrinologie, Universitätsklinikum Gießen und Marburg, Germany
| | - Thilo Hackert
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie Universitätsklinikum, Heidelberg, Germany
| | - Volker Heinemann
- Medizinische Klinik und Poliklinik III, Klinikum der Universität München-Campus Grosshadern, München, Germany
| | | | - Marianne Sinn
- Universitätsklinikum Hamburg-Eppendorf Medizinische Klinik und Poliklinik II Onkologie Hämatologie, Hamburg, Germany
| | | | | | - Waldemar Uhl
- Allgemein- und Viszeralchirurgie, St Josef-Hospital, Bochum, Germany
| |
Collapse
|
39
|
Li Q, Song Z, Zhang D, Li X, Liu Q, Yu J, Li Z, Zhang J, Ren X, Wen Y, Tang Z. Feasibility of a CT-based lymph node radiomics nomogram in detecting lymph node metastasis in PDAC patients. Front Oncol 2022; 12:992906. [PMID: 36276058 PMCID: PMC9579427 DOI: 10.3389/fonc.2022.992906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/20/2022] [Indexed: 12/02/2022] Open
Abstract
Objectives To investigate the potential value of a contrast enhanced computed tomography (CECT)-based radiological-radiomics nomogram combining a lymph node (LN) radiomics signature and LNs’ radiological features for preoperative detection of LN metastasis in patients with pancreatic ductal adenocarcinoma (PDAC). Materials and methods In this retrospective study, 196 LNs in 61 PDAC patients were enrolled and divided into the training (137 LNs) and validation (59 LNs) cohorts. Radiomic features were extracted from portal venous phase images of LNs. The least absolute shrinkage and selection operator (LASSO) regression algorithm with 10-fold cross-validation was used to select optimal features to determine the radiomics score (Rad-score). The radiological-radiomics nomogram was developed by using significant predictors of LN metastasis by multivariate logistic regression (LR) analysis in the training cohort and validated in the validation cohort independently. Its diagnostic performance was assessed by receiver operating characteristic curve (ROC), decision curve (DCA) and calibration curve analyses. Results The radiological model, including LN size, and margin and enhancement pattern (three significant predictors), exhibited areas under the curves (AUCs) of 0.831 and 0.756 in the training and validation cohorts, respectively. Nine radiomic features were used to construct a radiomics model, which showed AUCs of 0.879 and 0.804 in the training and validation cohorts, respectively. The radiological-radiomics nomogram, which incorporated the LN Rad-score and the three LNs’ radiological features, performed better than the Rad-score and radiological models individually, with AUCs of 0.937 and 0.851 in the training and validation cohorts, respectively. Calibration curve analysis and DCA revealed that the radiological-radiomics nomogram showed satisfactory consistency and the highest net benefit for preoperative diagnosis of LN metastasis. Conclusions The CT-based LN radiological-radiomics nomogram may serve as a valid and convenient computer-aided tool for personalized risk assessment of LN metastasis and help clinicians make appropriate clinical decisions for PADC patients.
Collapse
Affiliation(s)
- Qian Li
- Department of Radiology, Chongqing Medical University, Chongqing, China
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing, China
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Zuhua Song
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Dan Zhang
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Xiaojiao Li
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Qian Liu
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Jiayi Yu
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Zongwen Li
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Jiayan Zhang
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Xiaofang Ren
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Youjia Wen
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Zhuoyue Tang
- Department of Radiology, Chongqing Medical University, Chongqing, China
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing, China
- Department of Radiology, Chongqing General Hospital, Chongqing, China
- *Correspondence: Zhuoyue Tang,
| |
Collapse
|
40
|
Vivarelli M, Mocchegiani F, Nicolini D, Vecchi A, Conte G, Dalla Bona E, Rossi R, Benedetti Cacciaguerra A. Neoadjuvant Treatment in Resectable Pancreatic Cancer. Is It Time for Pushing on It? Front Oncol 2022; 12:914203. [PMID: 35712487 PMCID: PMC9195424 DOI: 10.3389/fonc.2022.914203] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic resection still represents the only curative option for patients affected by pancreatic ductal adenocarcinoma (PDAC). However, the association with modern chemotherapy regimens is a key factor in improving the inauspicious oncological outcome. The benefit of neoadjuvant treatment (NAT) for borderline resectable/locally advanced PDAC has been demonstrated; this evidence raises the question of whether even resectable PDAC should undergo NAT rather than upfront surgery. NAT may avoid futile surgery because of undetected distant metastases or aggressive tumor biology, providing more effective systemic control of the disease, which is hampered when adjuvant chemotherapy is delayed or precluded. However, recent data show controversial results regarding the efficacy and safety of NAT in resectable PDAC compared to upfront surgery. Although several prospective studies and meta-analyses indicate better oncologic outcomes after NAT, there are some biases, such as the methodological approaches used to capture the events of interest, which could make these results hardly reproducible. For instance, per-protocol studies, considering only the postoperative outcomes, tend to overestimate the performance of NAT by excluding patients who will never be suitable for surgery due to the development of chemotoxicity or tumor progression. To draw reliable conclusions, the studies should capture the events of interest of both strategies (NAT/upfront surgery) from the time of allocation to a specific treatment in an intention-to-treat fashion. This critical review highlights the current literature data concerning the use of NAT in resectable PDAC, summarizing the results of high-quality studies and focusing on the methodological issues of the most recent pieces of evidence.
Collapse
|
41
|
Keane F, Greally M, Horgan A, Duffy K, Lowery M, Martin P, Grogan L, Osman N, Power DG, Nasim S, O'Reilly EM, Leonard G. Consensus statement on the surveillance of patients with gastrointestinal malignancies. Ir J Med Sci 2022; 192:575-589. [PMID: 35616834 DOI: 10.1007/s11845-022-02987-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/16/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Improvements in early detection, screening and treatment of cancer have resulted in a significant improvement in cancer mortality and an increase in the number of cancer survivors globally. Accordingly, a significant rise in the number of cancer survivors in Ireland has been observed. The surveillance of survivors of gastrointestinal malignancies in Ireland is heterogeneous and represents an unmet need for standardisation. AIMS There are currently no national guidelines in Ireland to guide follow-up practices for these patients. The aim of this study was to establish homogeneity nationally with respect to follow-up of these patients by medical oncologists. METHODS/RESULTS A consensus group consisting of Irish oncologists with an interest in gastrointestinal malignancies was created to address this issue, and determined that it would be reasonable to adopt the NCCN guidelines for this purpose, but that this recommendation would not be prescriptive, and should be individualised to each patient. CONCLUSION We hope that this initiative may help to homogenise survivorship practices in this cohort of Irish patients, and may support the implementation of survivorship initiatives by the National Cancer Control Programme (NCCP).
Collapse
Affiliation(s)
- Fergus Keane
- University Hospital Galway, Newcastle Road, Galway, Ireland.
| | | | - Anne Horgan
- University Hospital Waterford, Dunmore Road, Waterford, Ireland
| | - Karen Duffy
- Letterkenny University Hospital, Kilmacrennan Road, Leterkenny, Donegal, Ireland
| | - Maeve Lowery
- St. James' Hospital, James Street, Dublin, Dublin 8, Ireland
| | - Petra Martin
- Midland Regional Hospital, Arden Road, Tullamore, Co. Offaly, Ireland
| | - Liam Grogan
- Beaumont Hospital, Beaumont Road, Dublin 9, Ireland
| | - Nemer Osman
- University Hospital Limerick, Dooradoyle, Co. Limerick, Ireland
| | - Derek G Power
- Cork University Hospital, Wilton Road, Cork, Co. Cork, Ireland
| | - Saira Nasim
- Midland Regional Hospital, Arden Road, Tullamore, Co. Offaly, Ireland
| | | | | |
Collapse
|
42
|
Sharma S, Tapper WJ, Collins A, Hamady ZZR. Predicting Pancreatic Cancer in the UK Biobank Cohort Using Polygenic Risk Scores and Diabetes Mellitus. Gastroenterology 2022; 162:1665-1674.e2. [PMID: 35065983 DOI: 10.1053/j.gastro.2022.01.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 01/04/2022] [Accepted: 01/10/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Diabetes mellitus (DM) is known to be associated with pancreatic ductal adenocarcinoma (PDAC), particularly new-onset DM (NODM). Others have developed polygenic risk scores (PRS) associated with PDAC risk. We aimed to compare the performance of these PRS in an independent cohort to determine if they can discriminate between NODM and long-standing DM patients with PDAC. METHODS Cases (1042) and matched cancer-free controls (10,420) were drawn from the UK Biobank. Five PRS models were calculated using single nucleotide polymorphisms (SNPs) from previous studies (Nakatochi, Galeotti, Molina, Jia, and Rashkin) and a combination of these. Regression models were used to assess the association between PDAC and PRS adjusted for ancestry, smoking, DM, waist circumference, and family history of digestive cancer. Receiver operator characteristic curves and area under the curve metrics (AUC) were used to assess the performance of each PRS for classifying PDAC risk. RESULTS The combined PRS model achieved the highest AUC (0.605), and significantly improved a clinical risk model in this cohort (AUC = 0.83; P = .0002). Individuals within the fifth quintile have a 2.74-fold increased risk of developing PDAC vs those in the first quintile (P < .001), and have a 3.05-fold increased risk of developing PDAC if they have DM vs those without DM (P < .001). The positive predictive value was 11.9% in participants without DM, 23.9% with long-standing DM, and 86.7% with NODM. CONCLUSIONS The PDAC-related common genetic variants are more strongly associated with DM. This PRS has the potential for targeting individuals with NODM for PDAC secondary screening measures.
Collapse
Affiliation(s)
- Shreya Sharma
- University of Southampton, Human Development and Health, Southampton, United Kingdom
| | - William J Tapper
- University of Southampton, Human Development and Health, Southampton, United Kingdom
| | - Andrew Collins
- University of Southampton, Genetic Epidemiology and Bioinformatics Research Group, Human Development and Health, Southampton, United Kingdom
| | - Zaed Z R Hamady
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.
| |
Collapse
|
43
|
Phase I Trial of nab-Paclitaxel Administered Concurrently With Radiotherapy in Patients With Locally Advanced Inoperable Pancreatic Adenocarcinoma. Pancreas 2022; 51:490-495. [PMID: 35849065 DOI: 10.1097/mpa.0000000000002065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Nab-paclitaxel has radiosensitizing antitumor efficacy in pancreatic cancer. We aimed to establish maximum tolerated dose (MTD) of nab-paclitaxel with radiotherapy in unresectable locally advanced pancreatic cancer. METHODS In a phase I dose escalation trial patients received weekly nab-paclitaxel for 6 weeks with external beam radiotherapy (EBRT). 3 + 3 design was used with nab-paclitaxel doses: 25 mg/m 2 (cohort 1), 50 mg/m 2 (cohort 2), 75 mg/m 2 (cohort 3), and 100 mg/m 2 (cohort 4). Primary endpoint was MTD. Secondary objectives were progression-free survival and overall survival. RESULTS Fourteen patients were recruited. Median age was 69 years (range, 40-86). Grade 1/2 toxicities were nausea (93%), vomiting (54%), diarrhea (57%), and fatigue (69%). There were no dose limiting toxicities (DLT) in cohorts 1 to 3. In cohort 4, DLTs of febrile neutropenia and enterocolitis were observed in patient 1. Subsequent DLT of febrile neutropenia and enterocolitis occurred in patient 5 in the expanded cohort. Following chemoradiotherapy median progression-free survival was 4.7 months (95% confidence interval, 2.5-27.5) and median overall survival was 10.8 months (95% confidence interval, 6.37-25.2). CONCLUSIONS Nab-paclitaxel and EBRT was well-tolerated at doses below 100 mg/m 2 . The MTD and recommended phase II study dose for nab-paclitaxel with EBRT is 75 mg/m 2 in this disease.
Collapse
|
44
|
Daamen LA, Groot VP, Besselink MG, Bosscha K, Busch OR, Cirkel GA, van Dam RM, Festen S, Groot Koerkamp B, Haj Mohammad N, van der Harst E, de Hingh IHJT, Intven MPW, Kazemier G, Los M, Meijer GJ, de Meijer VE, Nieuwenhuijs VB, Pranger BK, Raicu MG, Schreinemakers JMJ, Stommel MWJ, Verdonk RC, Verkooijen HM, Molenaar IQ, van Santvoort HC. Detection, Treatment, and Survival of Pancreatic Cancer Recurrence in the Netherlands: A Nationwide Analysis. Ann Surg 2022; 275:769-775. [PMID: 32773631 DOI: 10.1097/sla.0000000000004093] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To evaluate whether detection of recurrent pancreatic ductal adenocarcinoma (PDAC) in an early, asymptomatic stage increases the number of patients receiving additional treatment, subsequently improving survival. SUMMARY OF BACKGROUND DATA International guidelines disagree on the value of standardized postoperative surveillance for early detection and treatment of PDAC recurrence. METHODS A nationwide, observational cohort study was performed including all patients who underwent PDAC resection (2014-2016). Prospective baseline and perioperative data were retrieved from the Dutch Pancreatic Cancer Audit. Data on follow-up, treatment, and survival were collected retrospectively. Overall survival (OS) was evaluated using multivariable Cox regression analysis, before and after propensity-score matching, stratified for patients with symptomatic and asymptomatic recurrence. RESULTS Eight hundred thirty-six patients with a median follow-up of 37 months (interquartile range 30-48) were analyzed. Of those, 670 patients (80%) developed PDAC recurrence after a median follow-up of 10 months (interquartile range 5-17). Additional treatment was performed in 159/511 patients (31%) with symptomatic recurrence versus 77/159 (48%) asymptomatic patients (P < 0.001). After propensity-score matching on lymph node ratio, adjuvant therapy, disease-free survival, and recurrence site, additional treatment was independently associated with improved OS for both symptomatic patients [hazard ratio 0.53 (95% confidence interval 0.42-0.67); P < 0.001] and asymptomatic patients [hazard ratio 0.45 (95% confidence interval 0.29-0.70); P < 0.001]. CONCLUSIONS Additional treatment of PDAC recurrence was independently associated with improved OS, with asymptomatic patients having a higher probability to receive recurrence treatment. Therefore, standardized postoperative surveillance aiming to detect PDAC recurrence before the onset of symptoms has the potential to improve survival. This provides a rationale for prospective studies on standardized surveillance after PDAC resection.
Collapse
Affiliation(s)
- Lois A Daamen
- Department of Surgery, UMC Utrecht Cancer Center, Utrecht University, Utrecht, the Netherlands
- Department of Radiation Oncology, UMC Utrecht Cancer Center, Utrecht University, Utrecht, the Netherlands
| | - Vincent P Groot
- Department of Surgery, UMC Utrecht Cancer Center, Utrecht University, Utrecht, the Netherlands
| | - Marc G Besselink
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Koop Bosscha
- Department of Surgery, Jeroen Bosch Hospital, Den Bosch, the Netherlands
| | - Olivier R Busch
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Geert A Cirkel
- Department of Medical Oncology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Utrecht University, the Netherlands
- Department of Medical Oncology, Meander Medical Center, Amersfoort, the Netherlands
| | - Ronald M van Dam
- Department of Surgery, Maastricht UMC+, Maastricht, the Netherlands
| | | | | | - Nadia Haj Mohammad
- Department of Medical Oncology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Utrecht University, the Netherlands
| | | | | | - Martijn P W Intven
- Department of Radiation Oncology, UMC Utrecht Cancer Center, Utrecht University, Utrecht, the Netherlands
| | - Geert Kazemier
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| | - Maartje Los
- Department of Medical Oncology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Utrecht University, the Netherlands
| | - Gert J Meijer
- Department of Radiation Oncology, UMC Utrecht Cancer Center, Utrecht University, Utrecht, the Netherlands
| | - Vincent E de Meijer
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Bobby K Pranger
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mihaela G Raicu
- Department of Pathology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, the Netherlands
| | | | - Martijn W J Stommel
- Department of Surgery, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Robert C Verdonk
- Department of Gastroenterology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, the Netherlands
| | - Helena M Verkooijen
- Imaging Division, University Medical Centre Utrecht; Utrecht University, Utrecht, the Netherlands
| | - Izaak Quintus Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Utrecht University, the Netherlands
| | - Hjalmar C van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Utrecht University, the Netherlands
| |
Collapse
|
45
|
Soloff EV, Al-Hawary MM, Desser TS, Fishman EK, Minter RM, Zins M. Imaging Assessment of Pancreatic Cancer Resectability After Neoadjuvant Therapy: AJR Expert Panel Narrative Review. AJR Am J Roentgenol 2022; 218:570-581. [PMID: 34851713 DOI: 10.2214/ajr.21.26931] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite important innovations in the treatment of pancreatic ductal adenocarcinoma (PDAC), PDAC remains a disease with poor prognosis and high mortality. A key area for potential improvement in the management of PDAC, aside from earlier detection in patients with treatable disease, is the improved ability of imaging techniques to differentiate treatment response after neoadjuvant therapy (NAT) from worsening disease. It is well established that current imaging techniques cannot reliably make this distinction. This narrative review provides an update on the imaging assessment of pancreatic cancer resectability after NAT. Current definitions of borderline resectable PDAC, as well as implications for determining likely patient benefit from NAT, are described. Challenges associated with PDAC pathologic evaluation and surgical decision making that are of relevance to radiologists are discussed. Also explored are the specific limitations of imaging in differentiating the response after NAT from stable or worsening disease, including issues relating to protocol optimization, tumor size assessment, vascular assessment, and liver metastasis detection. The roles of MRI as well as PET and/or hybrid imaging are considered. Finally, a short PDAC reporting template is provided for use after NAT. The highlighted methods seek to improve radiologists' assessment of PDAC treatment response after NAT.
Collapse
Affiliation(s)
- Erik V Soloff
- Department of Radiology, University of Washington, Seattle, WA
| | - Mahmoud M Al-Hawary
- Department of Radiology and Internal Medicine, Michigan Medicine, Ann Arbor, MI
| | - Terry S Desser
- Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Elliot K Fishman
- Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD
| | - Rebecca M Minter
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Marc Zins
- Department of Radiology, Groupe Hospitalier Paris Saint Joseph, 185 Rue R Losserand, Paris 75014, France
| |
Collapse
|
46
|
Wang X, Xu W, Hu X, Yang X, Zhang M. The Prognostic Role of Glycemia in Patients With Pancreatic Carcinoma: A Systematic Review and Meta-Analysis. Front Oncol 2022; 12:780909. [PMID: 35223469 PMCID: PMC8866248 DOI: 10.3389/fonc.2022.780909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/03/2022] [Indexed: 12/14/2022] Open
Abstract
Background Fasting blood glucose and glycated hemoglobin (HbA1c) levels are associated with the risk of pancreatic cancer. Aim To examine the relationship between perioperative glucose and HbA1c levels and prognosis in patients with pancreatic cancer. Methods PubMed, Embase, and the Cochrane Library were queried for potentially eligible studies published up to May 2021. The exposures were perioperative fasting glucose and HbA1c levels. The primary outcome was survival. The secondary outcome was complications. All analyses were performed using the random-effects model. Results Ten studies (48,424 patients) were included. The pre-operative (HR=1.10, 95%CI: 0.89-1.35; I2 = 45.1%, Pheterogeneity=0.078) and postoperative (HR=1.19, 95%CI: 0.92-1.54; I2 = 67.9%, Pheterogeneity=0.001) blood glucose levels were not associated with the survival to pancreatic cancer. Similar results were observed for HbA1c (HR=1.09, 95%CI: 0.75-1.58; I2 = 64.2%, Pheterogeneity=0.039), fasting blood glucose (FBG)/HbA1c (HR=1.16, 95%CI: 0.67-1.68; I2 = 0.0%, Pheterogeneity=0.928), and FBG (HR=1.75, 95%CI: 0.81-3.75; I2 = 79.4%, Pheterogeneity=0.008). Pre-operative blood glucose levels were not associated with postoperative complications (OR=0.90, 95%CI: 0.52-1.56), but postoperative glucose levels were associated with postoperative complications (OR=3.06, 95%CI: 1.88-4.97; I2 = 0.0%, Pheterogeneity=0.619). Conclusion Blood glucose, FBG, and HbA1c levels are not associated with the survival of patients with pancreatic cancer. Postoperative blood glucose levels could predict postoperative complications.
Collapse
Affiliation(s)
- Xiaofang Wang
- Department of Clinical Oncology and Department of Hospice Care, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wanfeng Xu
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoru Hu
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xianghong Yang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mingming Zhang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
47
|
Li Q, Feng Z, Miao R, Liu X, Liu C, Liu Z. Prognosis and survival analysis of patients with pancreatic cancer: retrospective experience of a single institution. World J Surg Oncol 2022; 20:11. [PMID: 34996486 PMCID: PMC8742338 DOI: 10.1186/s12957-021-02478-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 12/07/2021] [Indexed: 12/15/2022] Open
Abstract
Background The overall survival of patients with pancreatic cancer is extremely low. Despite multiple large-scale studies, identification of predictors of patient survival remains challenging. This study aimed to investigate the prognostic factors for pancreatic cancer. Methods The clinical data of 625 patients with pancreatic cancer treated at Shengjing Hospital of China Medical University from January 2013 to December 2017 were collected. Results Of 625 patients, 569 were followed from 1 to 75 months. The median overall survival was 9.3 months. The overall 1-, 3-, and 5-year survival rates were 37.8%, 15.1%, and 10.5%, respectively. Cox proportional hazards model indicated that baseline carbohydrate antigen 199 level, neutrophil-lymphocyte ratio, operative procedure, lymph node metastasis, number of distant organ metastasis, and postoperative adjuvant chemotherapy were independent prognostic factors of patients with pancreatic cancer. Baseline carbohydrate antigen 199 level, degree of weight loss, operative procedure, lymph node metastasis, number of distant organ metastasis, and postoperative adjuvant chemotherapy were independent prognostic factors of pancreatic head cancer subgroup. Baseline carbohydrate antigen 199 level, carcinoembryonic antigen level, total bilirubin level, neutrophil-lymphocyte ratio, peripancreatic invasion, number of distant organ metastasis, and postoperative adjuvant chemotherapy were independent prognostic factors of the pancreatic body/tail cancer subgroup. Conclusions Higher carbohydrate antigen 199 levels, neutrophil-lymphocyte ratio, lymph node metastasis and distant organ metastasis predict a poor prognosis in patients with pancreatic cancer. Early detection, early radical surgery and adjuvant chemotherapy are needed to improve prognosis for this deadly disease.
Collapse
Affiliation(s)
- Qi Li
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, China
| | - Zijian Feng
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, China
| | - Ruyi Miao
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, China
| | - Xun Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, China
| | - Chenxi Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, China
| | - Zhen Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, China.
| |
Collapse
|
48
|
Irfan A, Rose JB, Wang TN, Vickers SM, Dudeja V, Gbolahan O, Reddy S. The Fate of Resectable Pancreatic Adenocarcinoma After Neoadjuvant Chemotherapy. Pancreas 2022; 51:100-105. [PMID: 35195602 DOI: 10.1097/mpa.0000000000001972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Pancreatic cancer continues to be a major cause of cancer-related mortality. There has been a greater implementation of up-front chemotherapy for pancreatic adenocarcinoma patients. Although there are many theoretical benefits to neoadjuvant chemotherapy, its clinical impact is uncertain. We sought to understand the outcomes of patients with resectable and borderline-resectable pancreatic adenocarcinoma who underwent neoadjuvant chemotherapy. METHODS Patients were collected in a secure database from September 2018 to May 2020. Patients were excluded if they presented with locally advanced or metastatic disease, inability to complete chemotherapy, or if they were not a surgical candidate. RESULTS Sixty-six patients with resectable disease underwent chemotherapy. Folinic acid/5-fluorouracil/irinotecan/oxaliplatin was used in 41 patients (62.1%) and gemcitabine-based regimens in 28 patients (42.4%, greater than 100% as some patients underwent both regimens). After restaging, 47 patients (71.2%) were thought to have resectable disease. Of these patients, 36 have been successfully resected to date. Metastatic disease was found in 12 patients (18.2%) and 6 patients (9.1%) had locally advanced disease. CONCLUSIONS Most patients with resectable pancreatic cancer are resected after neoadjuvant chemotherapy, but a subset will develop local or distant progression. Further studies will be needed to determine which patients will progress locally and may benefit from an up-front surgical approach.
Collapse
Affiliation(s)
| | | | | | | | | | - Olumide Gbolahan
- Division of Hematology Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | | |
Collapse
|
49
|
Rigiroli F, Hoye J, Lerebours R, Lafata KJ, Li C, Meyer M, Lyu P, Ding Y, Schwartz FR, Mettu NB, Zani S, Luo S, Morgan DE, Samei E, Marin D. CT Radiomic Features of Superior Mesenteric Artery Involvement in Pancreatic Ductal Adenocarcinoma: A Pilot Study. Radiology 2021; 301:610-622. [PMID: 34491129 PMCID: PMC9899097 DOI: 10.1148/radiol.2021210699] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Current imaging methods for prediction of complete margin resection (R0) in patients with pancreatic ductal adenocarcinoma (PDAC) are not reliable. Purpose To investigate whether tumor-related and perivascular CT radiomic features improve preoperative assessment of arterial involvement in patients with surgically proven PDAC. Materials and Methods This retrospective study included consecutive patients with PDAC who underwent surgery after preoperative CT between 2012 and 2019. A three-dimensional segmentation of PDAC and perivascular tissue surrounding the superior mesenteric artery (SMA) was performed on preoperative CT images with radiomic features extracted to characterize morphology, intensity, texture, and task-based spatial information. The reference standard was the pathologic SMA margin status of the surgical sample: SMA involved (tumor cells ≤1 mm from margin) versus SMA not involved (tumor cells >1 mm from margin). The preoperative assessment of SMA involvement by a fellowship-trained radiologist in multidisciplinary consensus was the comparison. High reproducibility (intraclass correlation coefficient, 0.7) and the Kolmogorov-Smirnov test were used to select features included in the logistic regression model. Results A total of 194 patients (median age, 66 years; interquartile range, 60-71 years; age range, 36-85 years; 99 men) were evaluated. Aside from surgery, 148 patients underwent neoadjuvant therapy. A total of 141 patients' samples did not involve SMA, whereas 53 involved SMA. A total of 1695 CT radiomic features were extracted. The model with five features (maximum hugging angle, maximum diameter, logarithm robust mean absolute deviation, minimum distance, square gray level co-occurrence matrix correlation) showed a better performance compared with the radiologist assessment (model vs radiologist area under the curve, 0.71 [95% CI: 0.62, 0.79] vs 0.54 [95% CI: 0.50, 0.59]; P < .001). The model showed a sensitivity of 62% (33 of 53 patients) (95% CI: 51, 77) and a specificity of 77% (108 of 141 patients) (95% CI: 60, 84). Conclusion A model based on tumor-related and perivascular CT radiomic features improved the detection of superior mesenteric artery involvement in patients with pancreatic ductal adenocarcinoma. © RSNA, 2021 Online supplemental material is available for this article. See also the editorial by Do and Kambadakone in this issue.
Collapse
Affiliation(s)
- Francesca Rigiroli
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| | - Jocelyn Hoye
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| | - Reginald Lerebours
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| | - Kyle J Lafata
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| | - Cai Li
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| | - Mathias Meyer
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| | - Peijie Lyu
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| | - Yuqin Ding
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| | - Fides R Schwartz
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| | - Niharika B Mettu
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| | - Sabino Zani
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| | - Sheng Luo
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| | - Desiree E Morgan
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| | - Ehsan Samei
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| | - Daniele Marin
- From the Departments of Radiology (F.R., K.J.L., M.M., P.L., Y.D., F.R.S., E.S., D.M.) and Radiation Oncology (K.J.L.), Duke University Medical Center, 2301 Erwin Rd, Box 3808, Durham, NC 27710; Multi-Dimensional Image Processing Laboratory, Duke Radiology, Duke University School of Medicine, Durham, NC (F.R., M.M., P.L., Y.D., F.R.S., D.M.); progettoDiventerò, Bracco Foundation, Milan, Italy (F.R.); Carl E. Ravin Advanced Imaging Laboratories (J.H., E.S.), Department of Biostatistics and Bioinformatics (R.L., S.L.), and Duke Electrical and Computer Engineering (K.J.L.), Duke University, Durham, NC; Department of Biostatistics, Yale University, New Haven, Conn (C.L.); Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany (M.M.); Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China (P.L.); Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China (Y.D.); Duke Cancer Center, Duke Health, Durham, NC (N.B.M., S.Z.); and Department of Radiology, University of Alabama at Birmingham, Birmingham, Ala (D.E.M.)
| |
Collapse
|
50
|
Moaven O, Clark CJ, Russell GB, Votanopoulos KI, Howerton R, Levine EA, Shen P. Optimal Adjuvant Treatment Approach After Upfront Resection of Pancreatic Cancer: Revisiting the Role of Radiation Based on Pathologic Features. Ann Surg 2021; 274:1058-1066. [PMID: 31913868 PMCID: PMC7335684 DOI: 10.1097/sla.0000000000003770] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To identify the survival benefit of different adjuvant approaches and factors influencing their efficacy after upfront resection of pancreatic ductal adenocarcinoma (PDAC). SUMMARY BACKGROUND DATA The optimal adjuvant approach for PDAC remains controversial. METHODS Patients from the National Cancer Database who underwent upfront PDAC resection from 2010 to 2014 were analyzed to determine clinical outcomes of different adjuvant treatment approaches, stratified according to pathologic characteristics. Factors associated with overall survival were identified with multivariable logistic regression and Cox proportional hazards were used to compare overall survival of different treatment approaches in the whole cohort, and propensity score matched groups. RESULTS We included 16,709 patients who underwent upfront resection of PDAC. On multivariable analysis, tumor size, grade, positive margin, nodal involvement, lymphovascular invasion (LVI), stage, lymph node ratio, not receiving chemotherapy, and/or radiation were predictors for worse survival. In the presence of at least 1 high-risk pathologic feature (nodal or margin involvement or LVI) chemotherapy with subsequent radiation provided the most significant survival benefit (median survivals: 24.8 vs 21.0 mo for adjuvant chemotherapy; HR = 0.81; 95% CI: 0.77-0.86; P < 0.001 in propensity score matching). The addition of radiation to adjuvant chemotherapy did not significantly improve overall survival in those with no high-risk pathologic features (median survivals: 54.6 vs 42.7 mo for adjuvant chemotherapy; HR=0.90; 95% CI: 0.75-1.08; P = 0.25 in propensity score matching). CONCLUSIONS In the presence of any high-risk pathologic features (nodal or margin involvement or LVI), adjuvant chemotherapy followed by radiation provides a better survival advantage over chemotherapy alone after upfront resection of PDAC.
Collapse
Affiliation(s)
- Omeed Moaven
- Division of Surgical Oncology, Department of Surgery, Wake Forest University, Winston Salem, NC
| | - Clancy J. Clark
- Division of Surgical Oncology, Department of Surgery, Wake Forest University, Winston Salem, NC
| | - Gregory B. Russell
- Department of Biostatistics and Data Science, Wake Forest University, Winston Salem, NC
| | | | - Russell Howerton
- Division of Surgical Oncology, Department of Surgery, Wake Forest University, Winston Salem, NC
| | - Edward A. Levine
- Division of Surgical Oncology, Department of Surgery, Wake Forest University, Winston Salem, NC
| | - Perry Shen
- Division of Surgical Oncology, Department of Surgery, Wake Forest University, Winston Salem, NC
| |
Collapse
|