1
|
Zhou SQ, Wan P, Zhang S, Ren Y, Li HT, Ke QH. Programmed cell death 1 inhibitor sintilimab plus S-1 and gemcitabine for liver metastatic pancreatic ductal adenocarcinoma. World J Clin Oncol 2025; 16:98079. [DOI: 10.5306/wjco.v16.i2.98079] [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: 06/17/2024] [Revised: 10/21/2024] [Accepted: 11/25/2024] [Indexed: 12/11/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with poor prognosis. When it metastasizes to the liver, treatment options become particularly limited and challenging. Current treatment options for liver metastatic PDAC are limited, and chemotherapy alone often proves insufficient. Immunotherapy, particularly programmed cell death 1 (PD-1) inhibitors like sintilimab, shows potential efficacy for various cancers but has limited reports on PDAC. This study compares the efficacy and safety of sintilimab plus S-1 and gemcitabine vs S-1 and gemcitabine alone in liver metastatic PDAC.
AIM To explore the feasibility and effectiveness of combined PD-1 inhibitor sintilimab and S-1 and gemcitabine (combination group) vs S-1 and gemcitabine used alone (chemotherapy group) for treating liver metastatic pancreatic adenocarcinoma.
METHODS Eligible patients were those with only liver metastatic PDAC, an Eastern Cooperative Oncology Group performance status of 0-1, adequate organ and marrow functions, and no prior anticancer therapy. Participants in the combination group received intravenous sintilimab 200 mg every 3 weeks, oral S-1 40 mg/m² twice daily on days 1-14 of a 21-day cycle, and intravenous gemcitabine 1000 mg/m² on days 1 and 8 of the same cycle for up to eight cycles or until disease progression, death, or unacceptable toxicity. Participants in the chemotherapy group received oral S-1 40 mg/m² twice daily on days 1-14 of a 21-day cycle and intravenous gemcitabine 1000 mg/m² on days 1 and 8 of the same cycle for up to eight cycles. Between June 2020 and December 2021, 66 participants were enrolled, with 32 receiving the combination treatment and 34 receiving chemotherapy alone.
RESULTS The group receiving the combined therapy exhibited a markedly prolonged median overall survival (18.8 months compared to 10.3 months, P < 0.05) and progression-free survival (9.6 months vs 5.4 months, P < 0.05). compared to the chemotherapy group. The incidence of severe adverse events did not differ significantly between the two groups (P > 0.05).
CONCLUSION The combination of PD-1 inhibitor sintilimab with S-1 and gemcitabine demonstrated effectiveness and safety for treating liver metastatic PDAC, meriting further investigation.
Collapse
Affiliation(s)
- Shi-Qiong Zhou
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Peng Wan
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Seng Zhang
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Yuan Ren
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Hong-Tao Li
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Qing-Hua Ke
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| |
Collapse
|
2
|
Blomstrand H, Bodarve M, Groth F, Naredi P, Sund M, Vilhav C, Green H, Björnsson B, Öhlund D, Lindblad S, Franklin O, Elander NO. Intratumoural expression of dihydropyrimidine dehydrogenase is an independent prognostic factor in resected pancreatic ductal adenocarcinoma treated with adjuvant gemcitabine. Oncol Lett 2025; 29:99. [PMID: 39703529 PMCID: PMC11656693 DOI: 10.3892/ol.2024.14845] [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: 09/16/2024] [Accepted: 11/14/2024] [Indexed: 12/21/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with a poor prognosis, and biomarkers to guide treatment decisions in PDAC are generally lacking. Intratumoural expression of dihydropyrimidine dehydrogenase (DPD) is a potential prognostic parameter in patients with PDAC undergoing surgical resection and postoperative chemotherapy. In the present study, DPD was analysed by immunohistochemistry of a tissue microarray platform including a real-world cohort of 495 patients with PDAC who had undergone resection with curative intent at any of three tertiary centres, including Northern, Western and Southeastern regions of Sweden, between 1993 and 2019. DPD level (high/low) was analysed and overall survival associations were assessed in treatment subgroups using a multivariate Cox regression model accounting for potential confounders. In patients who had not received adjuvant chemotherapy (n=182), the median overall survival time was 11.6 months (95% CI 9.6-13.5), compared with 28.8 months (25.0-32.6) among those who had (n=313; log-rank P<0.001). The most common type of chemotherapy was gemcitabine single agent (Gem, n=239) followed by gemcitabine plus capecitabine (GemCape, n=39). Tumour-Node-Metastasis (TNM) stage and DPD expression were statistically significant prognostic parameters in the Gem group (HR 1.19, 95% CI 1.01-1.41, P=0.036), with high expression of DPD linked with worse survival. In addition, tumour grade and TNM stage were statistically significant prognostic factors in the group that did not receive any chemotherapy (P≤0.001). No statistically significant parameters were identified in the GemCape group. Taken together, intratumoural expression of DPD may be considered a prognostic marker for patients with PDAC treated with adjuvant gemcitabine following surgical resection, with low expression levels predicting better survival. Further studies in larger cohorts of patients receiving multi-drug or non-gemcitabine based regimens are warranted.
Collapse
Affiliation(s)
- Hakon Blomstrand
- Division of Surgery, Orthopaedics and Oncology, Department of Biomedical and Clinical Sciences, Linköping University, SE-58185 Linköping, Sweden
- Department of Clinical Pathology, Linköping University Hospital, SE-58185 Linköping, Sweden
| | - Malin Bodarve
- Department of Clinical Pathology, Linköping University Hospital, SE-58185 Linköping, Sweden
| | - Fredrik Groth
- Department of Clinical Pathology, Linköping University Hospital, SE-58185 Linköping, Sweden
| | - Peter Naredi
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Malin Sund
- Department of Diagnostics and Intervention, Umeå University, SE-90187 Umeå, Sweden
- Department of Surgery, University of Helsinki and Helsinki University Hospital, 00014 Helsinki, Finland
| | - Caroline Vilhav
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Henrik Green
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, SE-58185 Linköping, Sweden
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping University, SE-58185 Linköping, Sweden
| | - Bergthor Björnsson
- Division of Surgery, Orthopaedics and Oncology, Department of Biomedical and Clinical Sciences, Linköping University, SE-58185 Linköping, Sweden
- Department of Surgery, Linköping University Hospital, SE-58185 Linköping, Sweden
| | - Daniel Öhlund
- Department of Diagnostics and Intervention, and Wallenberg Centre for Molecular Medicine, Umeå University, SE-90187 Umeå, Sweden
| | - Stina Lindblad
- Department of Diagnostics and Intervention, and Wallenberg Centre for Molecular Medicine, Umeå University, SE-90187 Umeå, Sweden
| | - Oskar Franklin
- Department of Diagnostics and Intervention, Umeå University, SE-90187 Umeå, Sweden
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Nils O. Elander
- Division of Surgery, Orthopaedics and Oncology, Department of Biomedical and Clinical Sciences, Linköping University, SE-58185 Linköping, Sweden
- Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool L7 8YA, United Kingdom
| |
Collapse
|
3
|
Madani SP, Mohseni A, Mirza-Aghazadeh-Attari M, Shahbazian H, Afyouni S, Borhani A, Zandieh G, Laheru D, Kamel IR. Role of volumetric tumor enhancement on CT in predicting overall survival in patients with unresectable pancreatic ductal adenocarcinoma. Clin Imaging 2025; 117:110365. [PMID: 39613522 DOI: 10.1016/j.clinimag.2024.110365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/06/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024]
Abstract
PURPOSE To assess the utility of volumetric tumor enhancement on CT to predict tumor treatment response and the overall survival (OS) of patients with PDAC undergoing FOLFIRINOX-based systemic chemotherapy. Additionally, we aim to explore the performance of a novel model that incorporates relevant volumetric CT-derived parameters to the established RECIST 1.1 in predicting both treatment response and OS. MATERIAL AND METHODS In this retrospective single-institution study, 127 patients with PDAC who received FOLFIRINOX neoadjuvant chemotherapy between December 2012 and November 2021 were included. Manual volumetric segmentation of the single largest tumor was performed on portal venous phase images. Total and enhancing tumor volumes were calculated. Response by RECIST 1.1 was compared to response by tumor volume and enhancing tumor volume on follow-up CT. RESULTS There was no association between overall survival and RECIST 1.1 (p-value = 0.284), volumetric RECIST (p-value = 0.402), and other volumetric CT variables, except for a percentage reduction in enhancing tumor volume (p-value = 0.043). Using univariate survival analysis for categorical thresholds defined by CART, the percentage change in enhancing tumor volume was associated with OS (p-value = 0.018). There was also a significant association between baseline enhancing tumor volume and OS (p-value <0.0001). Using these two categories, we defined a multivariable model associated with OS (p-value <0.0001). CONCLUSION Percentage reduction in enhancing tumor volume was related to OS in non-surgical PDAC patients treated with FOLFIRINOX chemotherapy and could potentially be incorporated into patient survival prediction models.
Collapse
Affiliation(s)
- Seyedeh Panid Madani
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Alireza Mohseni
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | | | - Haneyeh Shahbazian
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Shadi Afyouni
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Ali Borhani
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Ghazal Zandieh
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Daniel Laheru
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Ihab R Kamel
- Department of Radiology, University of Colorado, Aurora, CO, USA.
| |
Collapse
|
4
|
Habib JR, Kinny-Köster B, Javed AA, Zelga P, Saadat LV, Kim RC, Gorris M, Allegrini V, Watanabe S, Sharib J, Arcerito M, Kaiser J, Lafaro KJ, Tu M, Bhandre M, Shi C, Kim MP, Correa C, Daamen LA, Oberstein PE, Schmidt CM, Hanna NN, Allen P, Loos M, Shrikhande SV, Molenaar IQ, Frigerio I, Katz MHG, Soares KC, Miao Y, Del Chiaro M, He J, Hackert T, Salvia R, Büchler MW, Castillo CFD, Besselink MG, Marchegiani G, Wolfgang CL. Impact of Adjuvant Chemotherapy on Resected Intraductal Papillary Mucinous Neoplasm-Derived Pancreatic Cancer: Results From an International Multicenter Study. J Clin Oncol 2024; 42:4317-4326. [PMID: 39255450 DOI: 10.1200/jco.23.02313] [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: 10/24/2023] [Revised: 02/21/2024] [Accepted: 07/16/2024] [Indexed: 09/12/2024] Open
Abstract
PURPOSE The benefit of adjuvant therapy for intraductal papillary mucinous neoplasm (IPMN)-derived pancreatic ductal adenocarcinoma (PDAC) remains unclear because of severely limited evidence. Although biologically distinct entities, adjuvant therapy practices for IPMN-derived PDAC are largely founded on pancreatic intraepithelial neoplasia-derived PDAC. We aimed to evaluate the role of adjuvant chemotherapy in IPMN-derived PDAC. METHODS This international multicenter retrospective cohort study (2005-2018) was conceived at the Verona Evidence-Based Medicine meeting. Cox regressions were performed to identify risk-adjusted hazard ratios (HR) associated with overall survival (OS). Kaplan-Meier curves and log-rank tests were employed for survival analysis. Logistic regression was performed to identify factors motivating adjuvant chemotherapy administration. A decision tree was proposed and categorized patients into overtreated, undertreated, and optimally treated cohorts. RESULTS In 1,031 patients from 16 centers, nodal disease (HR, 2.88, P < .001) and elevated (≥37 to <200 µ/mL, HR, 1.44, P = .006) or markedly elevated (≥200 µ/mL, HR, 2.53, P < .001) carbohydrate antigen 19-9 (CA19-9) were associated with worse OS. Node-positive patients with elevated CA19-9 had an associated 34.4-month improvement in median OS (P = .047) after adjuvant chemotherapy while those with positive nodes and markedly elevated CA19-9 had an associated 12.6-month survival benefit (P < .001). Node-negative patients, regardless of CA19-9, did not have an associated benefit from adjuvant chemotherapy (all P > .05). Based on this model, we observed undertreatment in 18.1% and overtreatment in 61.2% of patients. Factors associated with chemotherapy administration included younger age, R1-margin, poorer differentiation, and nodal disease. CONCLUSION Almost half of patients with resected IPMN-derived PDAC may be overtreated or undertreated. In patients with node-negative disease or normal CA19-9, adjuvant chemotherapy is not associated with a survival benefit, whereas those with node-positive disease and elevated CA19-9 have an associated benefit from adjuvant chemotherapy. A decision tree was proposed. Randomized controlled trials are needed for validation.
Collapse
Affiliation(s)
- Joseph R Habib
- New York University Grossman School of Medicine, New York, NY
| | | | - Ammar A Javed
- New York University Grossman School of Medicine, New York, NY
| | | | - Lily V Saadat
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Rachel C Kim
- Indiana University School of Medicine, Indianapolis, IN
| | - Myrte Gorris
- Amsterdam University Medical Center, Amsterdam, the Netherlands
| | | | - Shuichi Watanabe
- University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO
| | | | | | - Jörg Kaiser
- Heidelberg University Hospital, Heidelberg, Germany
| | - Kelly J Lafaro
- The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Min Tu
- The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | | | | | - Michael P Kim
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Camilo Correa
- New York University Grossman School of Medicine, New York, NY
| | - Lois A Daamen
- University Medical Centre Utrecht, Utrecht, the Netherlands
| | | | - C Max Schmidt
- Indiana University School of Medicine, Indianapolis, IN
| | - Nader N Hanna
- University of Maryland Medical Center, Baltimore, MD
| | | | - Martin Loos
- Heidelberg University Hospital, Heidelberg, Germany
| | | | | | | | | | | | - Yi Miao
- The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Marco Del Chiaro
- University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO
| | - Jin He
- The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Thilo Hackert
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | |
Collapse
|
5
|
Gayibov E, Sychra T, Spálenková A, Souček P, Oliverius M. The use of patient-derived xenografts and patient-derived organoids in the search for new therapeutic regimens for pancreatic carcinoma. A review. Biomed Pharmacother 2024; 182:117750. [PMID: 39689516 DOI: 10.1016/j.biopha.2024.117750] [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: 09/06/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 12/19/2024] Open
Abstract
Patient-derived organoids (PDOs) and xenografts (PDXs) are powerful tools for personalized medicine in pancreatic cancer (PC) research. This study explores the complementary strengths of PDOs and PDXs in terms of practicality, genetic fidelity, cost, and labor considerations. Among other models like 2D cell cultures, spheroids, cancer-on-chip systems, cell line-derived xenografts (CDX), and genetically engineered mouse models (GEMMs), PDOs and PDXs uniquely balance genetic fidelity and personalized medicine potential, offering distinct advantages over the simplicity of 2D cultures and the advanced, but often resource-intensive, GEMMs and cancer-on-chip systems. PDOs excel in high-throughput drug screening due to their ease of use, lower cost, and shorter experimental timelines. However, they lack a complete tumor microenvironment. Conversely, PDXs offer a more complex microenvironment that closely reflects patient tumors, potentially leading to more clinically relevant results. Despite limitations in size, number of specimens, and engraftment success, PDXs demonstrate significant concordance with patient responses to treatment, highlighting their value in personalized medicine. Both models exhibit significant genetic fidelity, making them suitable for drug sensitivity testing. The choice between PDOs and PDXs depends on the research focus, resource availability, and desired level of microenvironment complexity. PDOs are advantageous for high-throughput screening of a diverse array of potential therapeutic agents due to their relative ease of culture and scalability. PDXs, on the other hand, offer a more physiologically relevant model, allowing for a comprehensive evaluation of drug efficacy and mechanisms of action.
Collapse
Affiliation(s)
- Emin Gayibov
- 3rd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tomáš Sychra
- 3rd Faculty of Medicine, Charles University, Prague, Czech Republic; Centre of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic; Department of General Surgery, 3rd Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| | - Alžběta Spálenková
- Centre of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Souček
- Centre of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| | - Martin Oliverius
- 3rd Faculty of Medicine, Charles University, Prague, Czech Republic; Department of General Surgery, 3rd Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic.
| |
Collapse
|
6
|
Agrawal S, Lee C, Pierce WF, Everhart E, King-Ducre A, Royce M, Osgood CL, Amiri-Kordestani L, Chiu HJ, Ricks TK, Pan L, Fourie Zirkelbach J, Charlab R, Pacanowski M, Kim T, Pazdur R, Kluetz PG, Gao JJ. FDA Approval Summary: Capecitabine Labeling Update under Project Renewal. Clin Cancer Res 2024; 30:5508-5514. [PMID: 39377782 DOI: 10.1158/1078-0432.ccr-24-1708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/20/2024] [Accepted: 10/07/2024] [Indexed: 10/09/2024]
Abstract
On December 14, 2022, the FDA approved revisions to the United States Prescribing Information (USPI) for capecitabine that revised existing indications and dosage regimens, added new indications and their recommended dosage regimens, revised safety information, updated the description of the risk of capecitabine in patients with dihydropyrimidine dehydrogenase deficiency, and edited other sections of the USPI to conform with FDA's current labeling guidance. These supplements were reviewed and approved under Project Renewal, a public health initiative established by the FDA's Oncology Center of Excellence that aims to update the prescribing information of certain older oncology drugs to ensure information is clinically meaningful and scientifically up to date. This article summarizes the FDA approach that supported revisions to the capecitabine USPI within the context of Project Renewal.
Collapse
Affiliation(s)
- Sundeep Agrawal
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
- Oncology Center of Excellence (OCE), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Clara Lee
- Oncology Center of Excellence (OCE), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - William F Pierce
- Oncology Center of Excellence (OCE), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Elizabeth Everhart
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Adriene King-Ducre
- Oncology Center of Excellence (OCE), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Melanie Royce
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Christy L Osgood
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Laleh Amiri-Kordestani
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Haw-Jyh Chiu
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Tiffany K Ricks
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Lili Pan
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Jeanne Fourie Zirkelbach
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Rosane Charlab
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Michael Pacanowski
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Tamy Kim
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
- Oncology Center of Excellence (OCE), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Richard Pazdur
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
- Oncology Center of Excellence (OCE), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Paul G Kluetz
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
- Oncology Center of Excellence (OCE), U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Jennifer J Gao
- Center for Drug Evaluation and Research (CDER), U.S. Food and Drug Administration, Silver Spring, Maryland
- Oncology Center of Excellence (OCE), U.S. Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
7
|
Biesma NC, Graus MUJE, Cirkel GA, Besselink MG, de Groot JWB, Koerkamp BG, Herbschleb KH, Los M, Verdonk RC, Wilmink JW, Cervantes A, Valle JW, Valkenburg-van Iersel LBJ, Froeling FEM, Molenaar IQ, Daamen LA, de Vos-Geelen J, van Santvoort HC. Perspectives of the medical oncologist regarding adjuvant chemotherapy for pancreatic cancer: An international expert survey and case vignette study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 51:109544. [PMID: 39689462 DOI: 10.1016/j.ejso.2024.109544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/19/2024] [Accepted: 12/10/2024] [Indexed: 12/19/2024]
Abstract
INTRODUCTION Adjuvant chemotherapy improves survival in patients with resected pancreatic ductal adenocarcinoma (PDAC). The decision to initiate chemotherapy involves both patient and physician factors, decision-specific criteria, and contextual considerations. This study aimed to assess medical oncologists' views on adjuvant chemotherapy following pancreatic resection for PDAC. METHODS An online survey and case vignette study were distributed to medical oncologists via the Dutch Pancreatic Cancer Group (DPCG), International Hepato-Pancreato-Biliary Association (IHPBA) and related networks. RESULTS A total of 91 oncologists from 14 countries participated, 46 % of whom treated more than 40 new PDAC patients annually, with a median experience of 15 years. Significant discrepancies were noted in their recommendations for adjuvant chemotherapy across case vignettes. In patients over 70, 17 % advised against chemotherapy, while 31 % said age was not a factor. Oncologists with less than 10 years of experience and those in non-academic settings were less likely to recommend adjuvant therapy. While 87 % agreed mFOLFIRINOX is the preferred adjuvant treatment, consensus on individual cases was lacking. The recommended interval between surgery and chemotherapy ranged from 3 to 26 weeks, with varying reasons for withholding treatment, primarily due to postoperative recovery and performance status. CONCLUSIONS Our study revealed substantial variation among oncologists in counseling on adjuvant chemotherapy after PDAC resection. This emphasizes the need for more patient involvement in decision-making and improving shared decision-making.
Collapse
Affiliation(s)
- N C Biesma
- University Medical Center Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Department of Surgery, the Netherlands.
| | - M U J E Graus
- Maastricht University Medical Center, Department of Internal Medicine, Division of Medical Oncology, GROW, Research Institute for Oncology & Reproduction, Maastricht, the Netherlands
| | - G A Cirkel
- University Medical Center Utrecht Cancer Center & St. Antonius Hospital Nieuwegein & Meander Medical Center Amersfoort, Regional Academic Cancer Center Utrecht, Department of Medical Oncology, the Netherlands
| | - M G Besselink
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands; Cancer Center Amsterdam, the Netherlands
| | - J W B de Groot
- Isala Oncology Center, Department of Medical Oncology, Zwolle, the Netherlands
| | - B Groot Koerkamp
- Erasmus Medical Center, Department of Surgery, Rotterdam, the Netherlands
| | - K H Herbschleb
- University Medical Center Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Department of Medical Oncology, the Netherlands
| | - M Los
- University Medical Center Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Department of Medical Oncology, the Netherlands
| | - R C Verdonk
- University Medical Center Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Department of Gastroenterology and Hepatology, the Netherlands
| | - J W Wilmink
- Cancer Center Amsterdam, the Netherlands; Amsterdam UMC, Location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands
| | - A Cervantes
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, University of Valencia, Spain
| | - J W Valle
- Cholangiocarcinoma Foundation, Herriman, Utah, USA; Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - L B J Valkenburg-van Iersel
- Maastricht University Medical Center, Department of Internal Medicine, Division of Medical Oncology, GROW, Research Institute for Oncology & Reproduction, Maastricht, the Netherlands
| | - F E M Froeling
- Dept. of Medical Oncology, Clinical Senior Lecturer and Honorary Consultant Medical Oncologist, University of Glasgow and Beatson West of Scotland Cancer Centre, United Kingdom
| | - I Q Molenaar
- University Medical Center Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Department of Surgery, the Netherlands
| | - L A Daamen
- University Medical Center Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Department of Surgery, the Netherlands; University Medical Center Utrecht Cancer Center, Division of Imaging & Oncology, Utrecht, the Netherlands
| | - J de Vos-Geelen
- Maastricht University Medical Center, Department of Internal Medicine, Division of Medical Oncology, GROW, Research Institute for Oncology & Reproduction, Maastricht, the Netherlands
| | - H C van Santvoort
- University Medical Center Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Department of Surgery, the Netherlands.
| |
Collapse
|
8
|
Haggstrom L, Chan WY, Nagrial A, Chantrill LA, Sim HW, Yip D, Chin V. Chemotherapy and radiotherapy for advanced pancreatic cancer. Cochrane Database Syst Rev 2024; 12:CD011044. [PMID: 39635901 PMCID: PMC11619003 DOI: 10.1002/14651858.cd011044.pub3] [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] [Indexed: 12/07/2024]
Abstract
BACKGROUND Pancreatic cancer (PC) is a lethal disease with few effective treatment options. Many anti-cancer therapies have been tested in the locally advanced and metastatic setting, with mixed results. This review synthesises all the randomised data available to help better inform patient and clinician decision-making. It updates the previous version of the review, published in 2018. OBJECTIVES To assess the effects of chemotherapy, radiotherapy, or both on overall survival, severe or life-threatening adverse events, and quality of life in people undergoing first-line treatment of advanced pancreatic cancer. SEARCH METHODS We searched for published and unpublished studies in CENTRAL, MEDLINE, Embase, and CANCERLIT, and handsearched various sources for additional studies. The latest search dates were in March and July 2023. SELECTION CRITERIA We included randomised controlled trials comparing chemotherapy, radiotherapy, or both with another intervention or best supportive care. Participants were required to have locally advanced, unresectable pancreatic cancer or metastatic pancreatic cancer not amenable to curative intent treatment. Histological confirmation was required. Trials were required to report overall survival. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. MAIN RESULTS We included 75 studies in the review and 51 in the meta-analysis (11,333 participants). We divided the studies into seven categories: any anti-cancer treatment versus best supportive care; various chemotherapy types versus gemcitabine; gemcitabine-based combinations versus gemcitabine alone; various chemotherapy combinations versus gemcitabine plus nab-paclitaxel; fluoropyrimidine-based studies; miscellaneous studies; and radiotherapy studies. In general, the included studies were at low risk for random sequence generation, detection bias, attrition bias, and reporting bias, at unclear risk for allocation concealment, and high risk for performance bias. Compared to best supportive care, chemotherapy likely results in little to no difference in overall survival (OS) (hazard ratio (HR) 1.08, 95% confidence interval (CI) 0.88 to 1.33; absolute risk of death at 12 months of 971 per 1000 versus 962 per 1000; 4 studies, 298 participants; moderate-certainty evidence). The adverse effects of chemotherapy and impacts on quality of life (QoL) were uncertain. Many of the chemotherapy regimens were outdated. Eight studies compared non-gemcitabine-based chemotherapy regimens to gemcitabine. These showed that 5-fluorouracil (5FU) likely reduces OS (HR 1.69, 95% CI 1.26 to 2.27; risk of death at 12 months of 914 per 1000 versus 767 per 1000; 1 study, 126 participants; moderate certainty), and grade 3/4 adverse events (QoL not reported). Fixed dose rate gemcitabine likely improves OS (HR 0.79, 95% CI 0.66 to 0.94; risk of death at 12 months of 683 per 1000 versus 767 per 1000; 2 studies, 644 participants; moderate certainty), and likely increase grade 3/4 adverse events (QoL not reported). FOLFIRINOX improves OS (HR 0.51, 95% CI 0.43 to 0.60; risk of death at 12 months of 524 per 1000 versus 767 per 1000; P < 0.001; 2 studies, 652 participants; high certainty), and delays deterioration in QoL, but increases grade 3/4 adverse events. Twenty-eight studies compared gemcitabine-based combinations to gemcitabine. Gemcitabine plus platinum may result in little to no difference in OS (HR 0.94, 95% CI 0.81 to 1.08; risk of death at 12 months of 745 per 1000 versus 767 per 1000; 6 studies, 1140 participants; low certainty), may increase grade 3/4 adverse events, and likely worsens QoL. Gemcitabine plus fluoropyrimidine improves OS (HR 0.88, 95% CI 0.81 to 0.95; risk of death at 12 months of 722 per 1000 versus 767 per 1000; 10 studies, 2718 participants; high certainty), likely increases grade 3/4 adverse events, and likely improves QoL. Gemcitabine plus topoisomerase inhibitors result in little to no difference in OS (HR 1.01, 95% CI 0.87 to 1.16; risk of death at 12 months of 770 per 1000 versus 767 per 1000; 3 studies, 839 participants; high certainty), likely increases grade 3/4 adverse events, and likely does not alter QoL. Gemcitabine plus taxane result in a large improvement in OS (HR 0.71, 95% CI 0.62 to 0.81; risk of death at 12 months of 644 per 1000 versus 767 per 1000; 2 studies, 986 participants; high certainty), and likely increases grade 3/4 adverse events and improves QoL. Nine studies compared chemotherapy combinations to gemcitabine plus nab-paclitaxel. Fluoropyrimidine-based combination regimens improve OS (HR 0.79, 95% CI 0.70 to 0.89; risk of death at 12 months of 542 per 1000 versus 628 per 1000; 6 studies, 1285 participants; high certainty). The treatment arms had distinct toxicity profiles, and there was little to no difference in QoL. Alternative schedules of gemcitabine plus nab-paclitaxel likely result in little to no difference in OS (HR 1.10, 95% CI 0.82 to 1.47; risk of death at 12 months of 663 per 1000 versus 628 per 1000; 2 studies, 367 participants; moderate certainty) or QoL, but may increase grade 3/4 adverse events. Four studies compared fluoropyrimidine-based combinations to fluoropyrimidines alone, with poor quality evidence. Fluoropyrimidine-based combinations are likely to result in little to no impact on OS (HR 0.84, 95% CI 0.61 to 1.15; risk of death at 12 months of 765 per 1000 versus 704 per 1000; P = 0.27; 4 studies, 491 participants; moderate certainty) versus fluoropyrimidines alone. The evidence suggests that there was little to no difference in grade 3/4 adverse events or QoL between the two groups. We included only one radiotherapy (iodine-125 brachytherapy) study with 165 participants. The evidence is very uncertain about the effect of radiotherapy on outcomes. AUTHORS' CONCLUSIONS Combination chemotherapy remains standard of care for metastatic pancreatic cancer. Both FOLFIRINOX and gemcitabine plus a taxane improve OS compared to gemcitabine alone. Furthermore, the evidence suggests that fluoropyrimidine-based combination chemotherapy regimens improve OS compared to gemcitabine plus nab-paclitaxel. The effects of radiotherapy were uncertain as only one low-quality trial was included. Selection of the most appropriate chemotherapy for individuals still remains unpersonalised, with clinicopathological stratification remaining elusive. Biomarker development is essential to assist in rationalising treatment selection for patients.
Collapse
Affiliation(s)
- Lucy Haggstrom
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- Medical Oncology, Illawarra Shoalhaven Local Health District, Wollongong, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Wei Yen Chan
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- Medical Oncology, Chris O'Brien Lifehouse, Sydney, Australia
| | - Adnan Nagrial
- The Crown Princess Mary Cancer Centre, Westmead, Australia
- Medical School, The University of Sydney, Sydney, Australia
| | - Lorraine A Chantrill
- Medical Oncology, Illawarra Shoalhaven Local Health District, Wollongong, Australia
- University of Wollongong, Wollongong, Australia
| | - Hao-Wen Sim
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Desmond Yip
- Department of Medical Oncology, The Canberra Hospital, Garran, Australia
- ANU Medical School, Australian National University, Acton, Australia
| | - Venessa Chin
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- Medical Oncology, Garvan Institute of Medical Research, Sydney, Australia
| |
Collapse
|
9
|
Palmer DH, Jackson R, Springfeld C, Ghaneh P, Rawcliffe C, Halloran CM, Faluyi O, Cunningham D, Wadsley J, Darby S, Meyer T, Gillmore R, Lind P, Glimelius B, Falk S, Ma YT, Middleton GW, Cummins S, Ross PJ, Wasan H, McDonald A, Crosby T, Hammel P, Borg D, Sothi S, Valle JW, Mehrabi A, Bailey P, Tjaden C, Michalski C, Hackert T, Büchler MW, Neoptolemos JP. Pancreatic Adenocarcinoma: Long-Term Outcomes of Adjuvant Therapy in the ESPAC4 Phase III Trial. J Clin Oncol 2024:JCO2401118. [PMID: 39637340 DOI: 10.1200/jco.24.01118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/08/2024] [Accepted: 10/03/2024] [Indexed: 12/07/2024] Open
Abstract
PURPOSE The ESPAC4 trial showed that adjuvant chemotherapy with gemcitabine plus capecitabine (GemCap) produced longer overall survival (OS) than gemcitabine monotherapy. Subsequently, the PRODIGE24-CCTG PA.6 trial showed even longer survival for modified fluorouracil, folinic acid, irinotecan, and oxaliplatin (mFOLFIRINOX) than gemcitabine but had more restrictive eligibility criteria. Our aim was to analyze the ESPAC4 survival on long-term follow-up. METHODS The OS of 732 ESPAC4 patients comparing 367 randomly assigned to gemcitabine and 365 to GemCap was previously reported after a median follow-up time of 43.2 months (95% CI, 39.7 to 45.5) and 458 deaths. Analysis was now carried out after a median follow-up of 104 months (101-108) and 566 deaths. RESULTS The median OS was 29.5 months (27.5-32.1) for all patients, 28.4 months (25.2-32.0) in the gemcitabine group and 31.6 months (26.5-38.0) in the GemCap group (hazard ratio [HR], 0.83 [0.71 to 0.98]; P = .031). R0 patients given gemcitabine had a median survival of 32.2 months (27.9-41.6) compared with 49.9 months (39.0-82.3) for those given GemCap (HR, 0.63 [0.47 to 0.84]; P = .002). Lymph node-negative patients had significantly higher 5 year OS rates on GemCap (59% [49%-71%]) than gemcitabine (53% [42%-66%]; HR, 0.63 [0.41 to 0.98]; P = .04) but not those with positive lymph nodes (P = .225). The OS advantage for GemCap was retained in the PRODIGE24 subgroup of 193 (26.4%) ESPAC4 patients not eligible for PRODIGE24 with a median survival of 20.7 (16.2-27.3) months in patients allocated to gemcitabine compared with 25.9 (22.3-30.2) months for ineligible patients allocated to GemCap (HR, 0.71 [95% CI, 0.52 to 0.98]; χ2log-rank-1df = 4.31; P = .038). CONCLUSION GemCap is a standard option for patients not eligible for mFOLFIRINOX. Exploratory evidence suggests that GemCap may be particularly efficacious in R0 patients and also in lymph node-negative patients.
Collapse
Affiliation(s)
| | | | - Christoph Springfeld
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Champalimaud Foundation, Lisbon, Portugal
| | - Paula Ghaneh
- University of Liverpool, Liverpool, United Kingdom
| | | | | | - Olusola Faluyi
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, United Kingdom
| | | | | | | | - Tim Meyer
- Department of Oncology, Royal Free Hospital and UCL Cancer Institute, University College London, London, United Kingdom
| | | | - Pehr Lind
- Department of Oncology, Stockholm Söder Hospital, Stockholm, Sweden
- Karolinska Institute, Stockholm, Sweden
| | - Bengt Glimelius
- Department of Immunology, Genetics and Pathology, University of Uppsala, Uppsala, Sweden
| | - Stephen Falk
- Bristol Cancer Institute, Bristol, United Kingdom
| | - Yuk Ting Ma
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | | | | | - Paul J Ross
- Guy's & St Thomas' and King's College Hospitals, London, United Kingdom
| | | | - Alec McDonald
- The Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Tom Crosby
- Velindre Cancer Centre, Cardiff, United Kingdom
| | - Pascal Hammel
- Hôpital Paul Brousse (APHP), Paris-Saclay University, Villejuif, France
| | - David Borg
- Skåne University Hospital, Lund, Sweden
- Department of Clinical Sciences Lund, Oncology and Therapeutic Pathology, Lund University, Lund, Sweden
| | - Sharmila Sothi
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
| | | | - Arianeb Mehrabi
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Bailey
- Champalimaud Foundation, Lisbon, Portugal
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christine Tjaden
- MRI TUM, Klinikum rechts der Isar of the Technical University of Munich, Munich, Germany
| | - Christoph Michalski
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Markus W Büchler
- Champalimaud Foundation, Lisbon, Portugal
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - John P Neoptolemos
- Champalimaud Foundation, Lisbon, Portugal
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
10
|
Crippa S, Malleo G, Langella S, Ricci C, Casciani F, Belfiori G, Galati S, Ingaldi C, Lionetto G, Ferrero A, Casadei R, Ercolani G, Salvia R, Falconi M, Cucchetti A. Cure Probabilities After Resection of Pancreatic Ductal Adenocarcinoma: A Multi-Institutional Analysis of 2554 Patients. Ann Surg 2024; 280:999-1005. [PMID: 38048334 DOI: 10.1097/sla.0000000000006166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
OBJECTIVE To assess the probability of being cured of pancreatic ductal adenocarcinoma (PDAC) by pancreatic surgery. BACKGROUND Statistical cure implies that a patient treated for a specific disease will have the same life expectancy as if he/she never had that disease. METHODS Patients who underwent pancreatic resection for PDAC between 2010 and 2021 were retrospectively identified using a multi-institutional database. A nonmixture statistical cure model was applied to compare disease-free survival to the survival expected for a matched general population. RESULTS Among 2554 patients, either in the setting of upfront (n=1691) or neoadjuvant strategy (n=863), the cure model showed that the probability that surgery would offer the same life expectancy (and tumor-free) as the matched general population was 20.4% (95% CI: 18.3, 22.5). Cure likelihood reached the 95% of certainty (time to cure) after 5.3 years (95% CI: 4.7, 6.0). A preoperative model was developed based on tumor stage at diagnosis ( P =0.001), radiologic size ( P =0.001), response to chemotherapy ( P =0.007), American Society of Anesthesiology class ( P =0.001), and preoperative Ca19-9 ( P =0.001). A postoperative model with the addition of surgery type ( P =0.015), pathologic size ( P =0.001), tumor grading ( P =0.001), resection margin ( P =0.001), positive lymph node ratio ( P =0.001), and the receipt of adjuvant therapy ( P =0.001) was also developed. CONCLUSIONS Patients operated for PDAC can achieve a life expectancy similar to that of the general population, and the likelihood of cure increases with the passage of recurrence-free time. An online calculator was developed and available at https://aicep.website/?cff-form=15 .
Collapse
Affiliation(s)
- Stefano Crippa
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Giuseppe Malleo
- Department of General and Pancreatic Surgery, Pancreas Institute, University of Verona Hospital Trust, Policlinico GB Rossi, Verona, Italy
| | - Serena Langella
- Department of General and Oncological Surgery, Mauriziano Hospital, Turin, Italy
| | - Claudio Ricci
- Division of Pancreatic Surgery, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Fabio Casciani
- Department of General and Pancreatic Surgery, Pancreas Institute, University of Verona Hospital Trust, Policlinico GB Rossi, Verona, Italy
| | - Giulio Belfiori
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Sara Galati
- Department of General and Oncological Surgery, Mauriziano Hospital, Turin, Italy
| | - Carlo Ingaldi
- Division of Pancreatic Surgery, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Gabriella Lionetto
- Department of General and Pancreatic Surgery, Pancreas Institute, University of Verona Hospital Trust, Policlinico GB Rossi, Verona, Italy
| | - Alessandro Ferrero
- Department of General and Oncological Surgery, Mauriziano Hospital, Turin, Italy
| | - Riccardo Casadei
- Division of Pancreatic Surgery, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Giorgio Ercolani
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
- Department of Surgery, Morgagni-Pierantoni Hospital, Forlì, Italy
| | - Roberto Salvia
- Department of General and Pancreatic Surgery, Pancreas Institute, University of Verona Hospital Trust, Policlinico GB Rossi, Verona, Italy
| | - Massimo Falconi
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandro Cucchetti
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
- Department of Surgery, Morgagni-Pierantoni Hospital, Forlì, Italy
| |
Collapse
|
11
|
Piper TB, Schaebel GH, Egeland C, Achiam MP, Burgdorf SK, Nerup N. Fluorescence-guided pancreatic surgery: A scoping review. Surgery 2024:108931. [PMID: 39613658 DOI: 10.1016/j.surg.2024.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 09/05/2024] [Accepted: 10/15/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Although fluorescence guidance during various surgical procedures has been shown to be safe and have possible better clinical outcomes than without the guidance, the use of fluorophores in pancreatic surgery is novel and not yet well described. This scoping review involved a systematic methodology of the currently available literature and aimed to illuminate the use of fluorophores in pancreatic surgery from a clinical view. METHODS The PRISMA and the PRISMA-ScR guidelines were used when appropriate and the following databases were searched: PubMed, Embase, Scopus, The Cochrane Collection, and Web of Science. Human original articles and case reports were included. Bias was assessed with the Newcastle-Ottawa Scale and the IDEAL framework was used for evaluation of surgical innovation. RESULTS A total of 5,565 search hits were screened, and 23 original articles and 24 case reports consisting of 754 patients met the inclusion criteria. The use of indocyanine green was both the most prominent and the most promising method for securing sufficient perfusion of neighboring organs, enhancing the detection and distinguishing of neuroendocrine tumors, and assisting in the identification of hepatic micrometastases. CONCLUSION The included studies were generally heterogenic, exploratory, and small. Indocyanine green was used in several ways, and it may add clinical value in different settings during pancreatic surgery. Tumor-targeted probes are a rapidly developing and promising field of research.
Collapse
Affiliation(s)
- Thomas B Piper
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | - Gustav H Schaebel
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Charlotte Egeland
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark. https://www.twitter.com/ChEgeland
| | - Michael P Achiam
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark. https://www.twitter.com/MichaelAchiam
| | - Stefan K Burgdorf
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Nikolaj Nerup
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark. https://www.twitter.com/nikolajnerup
| |
Collapse
|
12
|
Tushoski-Alemán GW, Crespin AJ, Oguejiofor CJ, Szymkiewicz DD, Herremans KM, Han S, Hughes SJ. Variability of quality-of-life measurements and reporting in randomised controlled trials of pancreatic cancer: a systematic review. BMJ Open 2024; 14:e083696. [PMID: 39551595 PMCID: PMC11574412 DOI: 10.1136/bmjopen-2023-083696] [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] [Indexed: 11/19/2024] Open
Abstract
OBJECTIVES This systematic review aims to evaluate the methodology used in pancreatic cancer (PC) randomised controlled trials (RCTs) measuring quality of life (QOL) and focuses on the type, frequency, survey compliance and duration of these assessments. DESIGN Systematic review of PC RCTs measuring QOL. DATA SOURCES A search of PubMed.gov and ClinicalTrials.gov was conducted for PC RCTs measuring QOL from inception to 21 March 2023. Only phase III RCTs were included. Studies were excluded if QOL was not measured, the study was phase I/II, in the second-line setting or unavailable in English. Data were independently extracted by two reviewers in a standardised fashion. PRIMARY AND SECONDARY OUTCOME MEASURES Primary outcomes included the type of QOL instrument used, the timing and frequency of assessments, methods of analysis and survey completion rates (SCRs) over time. Secondary outcomes included patient demographics, significant QOL improvements and the frequency of trials measuring QOL. RESULTS Out of 269 studies screened, 54 RCTs were identified, and 24 measured QOL (involving 11 229 patients). Instruments used included the EORTC QLQ-C30 (n=15), FACT-HEP (n=3), Spitzer-QOL-Index (n=2), EQ-5D (n=2), LASA (n=1) and FACT-PA (n=1). Most trials assessed QOL until disease progression or death (10/24), with 4-week intervals being the most common (7/24). SCRs were reported in 15/24 trials, with disease stage influencing SCRs over time. In trials with metastatic, locally advanced/metastatic, and resectable disease, the median times to reach a 50% response rate-defined as the point where the number of surveys completed was half of the enrolled participants-were 12.41 weeks (n=2), 14.14 weeks (n=10), and 54.2 weeks (n=3), respectively." Only 2/24 trials reported significant QOL improvements between treatment arms. Patient age was reported in all trials, while race/ethnicity was only reported in 4/24 trials. CONCLUSIONS Significant variability exists in the timing, methods and reporting of QOL assessments in PC trials. There is a need for further research to assess the implications of missing data and consider the temporality of QOL assessment in patients with advanced cancers and poor prognosis.
Collapse
Affiliation(s)
| | | | | | | | - Kelly M Herremans
- Department of Surgery, University of Florida, Gainesville, Florida, USA
| | - Song Han
- Department of Surgery, University of Florida, Gainesville, Florida, USA
| | - Steven J Hughes
- Department of Surgery, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
13
|
Miller ED, Ashman JB, Hawkins MA, Jethwa KR, Kim H, Sanford NN, Wojcieszynski AP, Chuong MD. The Dust Has Finally Settled, but Is the View Any Clearer? Int J Radiat Oncol Biol Phys 2024; 120:917-925. [PMID: 39424587 DOI: 10.1016/j.ijrobp.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 10/21/2024]
Affiliation(s)
- Eric D Miller
- Department of Radiation Oncology, Ohio State University Comprehensive Cancer Center, Columbus, Ohio.
| | | | - Maria A Hawkins
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Krishan R Jethwa
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Hyun Kim
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Nina N Sanford
- Department of Radiation Oncology, University of Texas Southwestern, Dallas, Texas
| | | | - Michael D Chuong
- Department of Radiation Oncology, Miami Cancer Institute, Miami, Florida
| |
Collapse
|
14
|
Hill CS, Parkinson R, Jaffee EM, Sugar E, Zheng L, Onners B, Weiss MJ, Wolfgang CL, Cameron JL, Pawlik TM, Rosati L, Le DT, Hacker-Prietz A, Lutz ER, Schulick R, Narang AK, Laheru DA, Herman JM. Phase 1 Study of Adjuvant Allogeneic Granulocyte-Macrophage Colony-Stimulating Factor-Transduced Pancreatic Tumor Cell Vaccine, Low-Dose Cyclophosphamide, and Stereotactic Body Radiation Therapy Followed by FOLFIRINOX in High-Risk Resected Pancreatic Ductal Adenocarcinoma. Int J Radiat Oncol Biol Phys 2024:S0360-3016(24)03561-2. [PMID: 39547453 DOI: 10.1016/j.ijrobp.2024.10.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/06/2024] [Accepted: 10/06/2024] [Indexed: 11/17/2024]
Abstract
PURPOSE Local and distant progression remains common following resection of resectable pancreatic ductal adenocarcinoma (PDAC) despite adjuvant multiagent chemotherapy. We report a prospective institutional phase 1 trial incorporating adjuvant GVAX vaccine, low-dose cyclophosphamide (Cy), and stereotactic body radiation therapy (SBRT) followed by FOLFIRINOX (FFX) among patients who underwent resection of high-risk PDAC. PATIENTS AND METHODS The study design was a modified 3+3. Cohort 1 received 5-fraction SBRT to 33 Gy to the tumor bed and 25 Gy to elective nodes followed by 6 cycles of full-dose FFX. After toxicity review, cohort 2 had SBRT and was switched to modified FFX (mFFX). Cohort 3 had 1 cycle of Cy/GVAX followed by SBRT, mFFX, and 4 cycles of maintenance Cy/GVAX with 6-month Cy/GVAX boosts until progression. RESULTS Nineteen patients were enrolled with a median follow-up of 36.2 months. To be eligible, patients were required to have close/positive margins (within ≤1 mm) (71%) and/or lymph node metastasis (79%). Overall, 63% of patients had both. In cohort 1, 67% of patients received 6 cycles of FFX; in cohort 2, 75% received 6 cycles of modified FFX. In cohort 3, 12 patients received the first dose of Cy/GVAX and SBRT with 10 individuals (83%) receiving 6 cycles of mFFX. Cohort 3 had acceptable levels of grade ≥3 thrombocytopenia, neutropenia, and diarrhea after 2 cycles of mFFX. Median overall survival (OS)/disease-free survival (DFS) for the overall cohort and cohort 3 was 36.2/18.2 months and 61.3/24.1 months, respectively. One- and 2-year OS for cohort 3 was 83%/75%, respectively. At the last follow-up (median = x), 5 patients were alive (42%) in cohort 3. CONCLUSIONS This is the first prospective trial to evaluate adjuvant GVAX, Cy, SBRT, and mFFX in resected PDAC patients with high-risk features. This combination regimen was well tolerated with limited toxicity and promising survival outcomes, warranting future studies to validate this regimen in the adjuvant setting.
Collapse
Affiliation(s)
- Colin S Hill
- Laura and Issac Perlmutter Cancer Center at New York University, Department of Radiation Oncology, New York University Grossman School of Medicine, New York, New York
| | - Rose Parkinson
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Cancer Convergence Institute and Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth M Jaffee
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Cancer Convergence Institute and Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth Sugar
- Division of Biostatistics and Bioinformatics, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Lei Zheng
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Cancer Convergence Institute and Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Beth Onners
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Cancer Convergence Institute and Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Matthew J Weiss
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Lake Success, New York, New York
| | - Christopher L Wolfgang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, New York, University Grossman School of Medicine, New York, New York
| | - John L Cameron
- Department of Surgery, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Cancer Convergence Institute and Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Timothy M Pawlik
- Department of Surgery, The Urban Meyer III and Shelley Meyer Chair for Cancer Research, The Ohio State University, Wexner Medical Center, Columbus, Ohio
| | - Lauren Rosati
- Department of Pediatrics, Heersink School of Medicine, University of Alabama, Birmingham, Alabama
| | - Dung T Le
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Cancer Convergence Institute and Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Amy Hacker-Prietz
- Department of Radiation Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Cancer Convergence Institute and Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Richard Schulick
- Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado; University of Colorado Cancer Center, Aurora, Colorado
| | - Amol K Narang
- Department of Radiation Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Cancer Convergence Institute and Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Daniel A Laheru
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Cancer Convergence Institute and Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Joseph M Herman
- Radiation Medicine, Zucker School of Medicine at Hofstra/Northwell, Lake Success, New York, New York..
| |
Collapse
|
15
|
Vaezi MA, Nekoufar S, Robati AK, Salimi V, Tavakoli-Yaraki M. Therapeutic potential of β-hydroxybutyrate in the management of pancreatic neoplasms: exploring novel diagnostic and treatment strategies. Lipids Health Dis 2024; 23:376. [PMID: 39543582 PMCID: PMC11562866 DOI: 10.1186/s12944-024-02368-7] [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: 08/22/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024] Open
Abstract
Pancreatic neoplasm, a highly aggressive and often fatal cancer, poses challenges due to late detection and nonspecific symptoms. Therefore, both early diagnosis and appropriate therapeutic approaches are necessary to augment the condition of these patients. Cancer cells undergo metabolic deregulation, which enables their proliferation, survival, and invasion. As a result, it is crucial to focus on the metabolic pathways in prevalent cancers and explore treatment strategies that target these pathways to control tumor growth effectively. This is particularly relevant in cancers like pancreatic cancer, which undergo numerous metabolic alterations. The ketogenic regimen, characterized by low carbohydrate and protein contents and high-fat sources, does not involve caloric restriction. This allows for the induction of ketogenesis and an increase in ketone bodies, while insulin and glucose levels remain low even after meals. This unique metabolic state may influence the tumor microenvironment. Given the lack of unanimous agreement on the precise role and mechanism of the ketogenic diet, this review aims to clarify the diagnostic value and accuracy of ketone bodies in various types of pancreatic tumors and explore the potential anti-cancer effects of the ketogenic diet when used alone or in conjunction with chemotherapy, also to determine the potential of the ketogenic diet to be used as adjuvant therapy. The outcomes of this study are instrumental in enhancing our understanding of the benefits and drawbacks associated with employing this diet for the management and diagnosis of pancreatic cancer.
Collapse
Affiliation(s)
- Mohammad Amin Vaezi
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Samira Nekoufar
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Ali Karami Robati
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Vahid Salimi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Tavakoli-Yaraki
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran.
- Finetech in Medicine Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Huang Y, Zhang H, Ding Q, Chen D, Zhang X, Weng S, Liu G. Comparison of multiple machine learning models for predicting prognosis of pancreatic ductal adenocarcinoma based on contrast-enhanced CT radiomics and clinical features. Front Oncol 2024; 14:1419297. [PMID: 39605884 PMCID: PMC11598923 DOI: 10.3389/fonc.2024.1419297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Objective The aim of this study was to evaluate the prognostic potential of combining clinical features and radiomics with multiple machine learning (ML) algorithms in pancreatic ductal adenocarcinoma (PDAC). Methods A total of 116 patients with PDAC who met the eligibility criteria were randomly assigned to a training or validation cohort. Seven ML algorithms, including Supervised Principal Components, stepwise Cox, Random Survival Forest, CoxBoost, Least absolute shrinkage and selection operation (Lasso), Ridge, and Elastic network, were integrated into 43 algorithm combinations. Forty-three radiomics models were constructed separately using radiomics features extracted from arterial phase (AP), venous phase (VP), and combined arterial and venous phase (AP+VP) images. The concordance index (C-index) of each model was calculated. The model with the highest mean C-index was identified as the best model for calculating the radiomics score (Radscore). Univariate and multivariate Cox analyses were used to identify independent prognostic indicators and create a clinical model for prognosis prediction. The multivariable Cox regression was used to combine Radscore with clinical features to create a combined model. The efficacy of the model was evaluated using the C-index, calibration curves, and decision curve analysis (DCA). Results The model based on the Lasso+StepCox[both] algorithm constructed using AP+VP radiomics features showed the best predictive ability among the 114 radiomics models. The C-indices of the model in the training and validation cohorts were 0.742 and 0.722, respectively. Based on the results of the univariate and multivariate Cox regression analyses, sex, Tumor-Node-Metastasis (TNM) stage, and systemic inflammation response index were included to build the clinical model. The combined model, incorporating three clinical factors and AP+VP-Radscore, achieved the highest C-indices of 0.764 and 0.746 in the training and validation cohorts, respectively. In terms of preoperative prognosis prediction for PDAC, the calibration curve and DCA showed that the combined model had a good consistency and greatest net benefit. Conclusion A combined model of clinical features and AP+VP-Radscore screened using multiple ML algorithms has an excellent ability to predict the prognosis of PDAC and may provide a noninvasive and effective method for clinical decision-making.
Collapse
Affiliation(s)
- Yue Huang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Han Zhang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Qingzhu Ding
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Dehua Chen
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xiang Zhang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Shangeng Weng
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Clinical Research Center for Hepatobiliary Pancreatic and Gastrointestinal Malignant Tumors Precise Treatment of Fujian, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Guozhong Liu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
17
|
Bryant JM, Nakashima J, Khatri VM, Sinnamon AJ, Denbo JW, Hodul P, Malafa M, Hoffe S, Frakes JM. The Evolving Role of Neoadjuvant Radiation Therapy in Pancreatic Adenocarcinoma. J Clin Med 2024; 13:6800. [PMID: 39597944 PMCID: PMC11594810 DOI: 10.3390/jcm13226800] [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: 10/12/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers. Surgical resection is the most reliable chance for cure, but high rates of positive margins and local failure persist. Neoadjuvant therapies (NAT), including chemotherapy and radiation therapy (RT), are being explored to improve surgical outcomes, particularly in borderline resectable (BRPC) and locally advanced pancreatic cancer (LAPC). This review aims to summarize the current landscape and future directions for neoadjuvant RT (NART) in PDAC. METHODS The review includes a detailed analysis of past and ongoing clinical trials investigating various NART approaches in PDAC, with an emphasis on different RT techniques, fractionation schemes, and their integration into multimodal treatment strategies. RESULTS Early evidence suggests that NART can improve resection margins and local control. However, recent trials, including the Alliance A021501 and LAP-07 trials, have failed to demonstrate significant survival benefits with the addition of RT to NAT. Nevertheless, nuances in trial design and execution continue to keep the question of NART open. Newer approaches, such as stereotactic magnetic resonance-guided adaptive radiation therapy (SMART), show promise in improving local control and survival, but further phase 3 trials are needed. CONCLUSIONS While NART has shown potential in improving local control in PDAC, its impact on overall survival remains unclear. Ongoing trials, particularly those utilizing advanced techniques like SMART, are critical in defining the role of RT in the neoadjuvant setting for PDAC. Collaboration across multidisciplinary teams is essential to optimize treatment strategies and trial outcomes.
Collapse
Affiliation(s)
- John Michael Bryant
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Justyn Nakashima
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Vaseem M. Khatri
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Andrew J. Sinnamon
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Jason W. Denbo
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Pamela Hodul
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Mokenge Malafa
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Sarah Hoffe
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Jessica M. Frakes
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
18
|
Maehira H, Mori H, Nitta N, Maekawa T, Nishina Y, Ishikawa H, Takebayashi K, Kaida S, Miyake T, Tani M. Clinical impact of the prognostic nutritional index and skeletal muscle index for the incompletion of adjuvant chemotherapy for pancreatic cancer. Asian J Surg 2024:S1015-9584(24)02484-9. [PMID: 39537486 DOI: 10.1016/j.asjsur.2024.10.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 09/26/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Adjuvant chemotherapy is a standard therapeutic option for resected pancreatic cancer. However, the risk factors for incompletion of adjuvant chemotherapy remain unclear. METHODS We retrospectively reviewed the medical records of 72 patients who underwent radical pancreatectomy and received S-1 adjuvant chemotherapy for pancreatic cancer. The patients were assigned to two groups according to their completion or incompletion of adjuvant chemotherapy. We compared the perioperative skeletal muscle mass index (SMI) and nutritional status using prognostic nutritional index (PNI) between the two groups. RESULTS The completion and incompletion groups included 46 (64 %) and 26 (36 %) patients, respectively. Overall survival was shorter in the incompletion group than in the completion group (median survival time, 20.2 months vs. 42.0 months; log-rank, p = 0.018). Decreasing rate of PNI (12.7 % vs. 0.2 %, p = 0.010) and decreasing rate of SMI (26.9 % vs. 12.5 %, p = 0.001) were significantly larger in the incompletion group than in the completion group. Multivariate analysis showed that decreasing rate of PNI (p = 0.016), decreasing rate of SMI (p = 0.013), and old age (p = 0.049) were independent risk factors for incompletion of S-1 adjuvant chemotherapy. Regarding the time-series variations, PNI improved from 1 to 3 months after pancreatectomy in the completion group (p = 0.006). Furthermore, the decreasing slope of SMI was stronger in the incompletion group. CONCLUSION Postoperative decrease of PNI and SMI is associated with the incompletion of S-1 adjuvant chemotherapy.
Collapse
Affiliation(s)
- Hiromitsu Maehira
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan.
| | - Haruki Mori
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Nobuhito Nitta
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Takeru Maekawa
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Yusuke Nishina
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Hajime Ishikawa
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| | | | - Sachiko Kaida
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Toru Miyake
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Masaji Tani
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| |
Collapse
|
19
|
An J, Kurilov R, Peccerella T, Bergmann F, Edderkaoui M, Lim A, Zhou X, Pfütze K, Schulz A, Wolf S, Hu K, Springfeld C, Mughal SS, Zezlina L, Fortunato F, Beyer G, Mayerle J, Roth S, Hulkkonen J, Merz D, Ei S, Mehrabi A, Loos M, Al-Saeedi M, Michalski CW, Büchler MW, Hackert T, Brors B, Pandol SJ, Bailey P, Neoptolemos JP. Metavert synergises with standard cytotoxics in human PDAC organoids and is associated with transcriptomic signatures of therapeutic response. Transl Oncol 2024; 49:102109. [PMID: 39217851 PMCID: PMC11402625 DOI: 10.1016/j.tranon.2024.102109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/18/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Despite some recent advances, pancreatic ductal adenocarcinoma (PDAC) remains a growing oncological challenge. New drugs capable of targeting more than one oncogenic pathway may be one way to improve patient outcomes. This study characterizes the effectiveness of Metavert a first-in-class dual inhibitor of GSK3-β and histone deacetylase in treating PDAC as a single agent or in combination with standard cytotoxics. METHODS Thirty-six Patient-Derived Organoids (hPDOs) characterised by RNASeq and whole exome sequencing were treated with Metavert alone or in combination with standard cytotoxics. Transcriptomic signatures (TS) representing sensitivity to Metavert alone or sensitivity to Metavert + irinotecan (IR) were evaluated in 47 patient samples, chemo-naïve in 26 and post-chemotherapy in 21 (gemcitabine=5; FOLFIRINOX=14, both=2) with companion multiplexed immunofluorescence and RNASeq data. RESULTS Metavert combined with gemcitabine, irinotecan, 5FU, oxaliplatin, and paclitaxel was synergistic in the hPDOs. Basal-subtype hPDOs were more sensitive to Metavert alone whereas the Metavert+IR combination exhibited synergy in Classical-subtype hPDOs with increased apoptosis and autophagy. hPDO-derived TS evaluated in PDAC tissues demonstrated that Metavert-TSHi samples were enriched for mRNA splicing and DNA repair processes; they were associated with Basal-like tissues but also with GATA6+ve-chemo-naïve samples and were higher following gemcitabine but not FOLFIRINOX treatment. In contrast, Metavert+IR-TSHI samples were enriched for TP53 pathways; they were associated with Classical-like pretreatment samples and with GATA6+ve/KRT17+ve hybrid cell types following FOLFIRINOX, but not gemcitabine treatment, and were unrelated to transcriptional subtypes. CONCLUSIONS Metavert as a single agent and in combination with irinotecan offers novel strategies for treating pancreatic cancer.
Collapse
Affiliation(s)
- Jingyu An
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Roma Kurilov
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Berliner Str. 41, Heidelberg 69120, Germany
| | - Teresa Peccerella
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Frank Bergmann
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Mouad Edderkaoui
- Department of Medicine, Cedars-Sinai Medical Center and University of California at Los Angeles, Thalians W204 8700 Beverly Blvd. Los Angeles, California CA 90048, United States
| | - Adrian Lim
- Department of Medicine, Cedars-Sinai Medical Center and University of California at Los Angeles, Thalians W204 8700 Beverly Blvd. Los Angeles, California CA 90048, United States
| | - Xu Zhou
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Katrin Pfütze
- Sample Processing Laboratory, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Angela Schulz
- NGS Core Facility, The German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan Wolf
- NGS Core Facility, The German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kai Hu
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Christoph Springfeld
- Department of Medical Oncology, National Center for Tumor Diseases, University Clinic Heidelberg, Heidelberg 69120, Germany
| | - Sadaf S Mughal
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Berliner Str. 41, Heidelberg 69120, Germany
| | - Lenart Zezlina
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Berliner Str. 41, Heidelberg 69120, Germany
| | - Franco Fortunato
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Georg Beyer
- Department of Internal Medicine II, Ludwig-Maximilians-University of Munich, Germany
| | - Julia Mayerle
- Department of Internal Medicine II, Ludwig-Maximilians-University of Munich, Germany
| | - Susanne Roth
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Johannes Hulkkonen
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Daniela Merz
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Shigenori Ei
- Department of Gastroenterological Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Arianeb Mehrabi
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Martin Loos
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Mohammed Al-Saeedi
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Christoph W Michalski
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Markus W Büchler
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany; Botton-Champalimaud Pancreatic Cancer Centre, Lisbon, Portugal
| | - Thilo Hackert
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany; Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Berliner Str. 41, Heidelberg 69120, Germany; German Cancer Consortium (DKTK), Core Center Heidelberg, Im Neuenheimer Feld 280, Heidelberg 69120, Germany; Medical Faculty and Faculty of Biosciences, Heidelberg University, Im Neuenheimer Feld 234, Heidelberg 69120, Germany; National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg 69120, Germany
| | - Stephen J Pandol
- Department of Medicine, Cedars-Sinai Medical Center and University of California at Los Angeles, Thalians W204 8700 Beverly Blvd. Los Angeles, California CA 90048, United States
| | - Peter Bailey
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Berliner Str. 41, Heidelberg 69120, Germany; Botton-Champalimaud Pancreatic Cancer Centre, Lisbon, Portugal.
| | - John P Neoptolemos
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany; Botton-Champalimaud Pancreatic Cancer Centre, Lisbon, Portugal.
| |
Collapse
|
20
|
Cerviño RH, Gómez N, Sahores A, Gouts A, González B, Shayo C, Davio C, Yaneff A. Flurbiprofen inhibits cAMP transport by MRP4/ABCC4 increasing the potency of gemcitabine treatment in PDAC cell models. Int J Biol Macromol 2024; 280:136386. [PMID: 39378921 DOI: 10.1016/j.ijbiomac.2024.136386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 10/10/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a highly malignant cancer with a grim prognosis due to its early metastasis and resistance to current chemotherapies, such as Gemcitabine (GEM). We have previously demonstrated that cAMP exclusion by MRP4 is critical for PDAC cell proliferation, establishing this transporter as a promising prognostic marker and therapeutic target. In search for novel therapeutic options to improve GEM efficacy, we conducted a drug repositioning screening to identify potential inhibitors of cAMP transport by MRP4. Several non-steroidal anti-inflammatory drugs (NSAIDs) can inhibit the transport of certain MRP4 substrates. In this study, we assessed the efficacy of sixteen NSAIDs in inhibiting cAMP transport mediated by MRP4, identifying seven potent inhibitors based on their IC50 values. The most potent inhibitors were further tested for their effect on cell proliferation and migration. Flurbiprofen emerged as the most potent inhibitor of both MRP4-mediated cAMP transport and cell proliferation. Overexpression of MRP4 in BxPC-3 cells significantly increased GEM resistance, and co-administration of flurbiprofen with GEM markedly enhanced the latter's potency inhibiting PDAC cells proliferation. These findings position flurbiprofen as a potent inhibitor of cAMP transport by MRP4 and a promising adjunctive therapy to enhance GEM effectiveness in PDAC treatment.
Collapse
Affiliation(s)
- Ramiro Héctor Cerviño
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Farmacia y Bioquímica, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Natalia Gómez
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Farmacia y Bioquímica, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Ana Sahores
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Farmacia y Bioquímica, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Agustín Gouts
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Farmacia y Bioquímica, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Betina González
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Farmacia y Bioquímica, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Carina Shayo
- Instituto de Biología y Medicina Experimental (Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Carlos Davio
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Farmacia y Bioquímica, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Agustín Yaneff
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Farmacia y Bioquímica, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
21
|
Qadir RMAB, Umair MB, Tariq UB, Ahmad A, Kiran W, Shahid MH. Unraveling Pancreatic Cancer: Epidemiology, Risk Factors, and Global Trends. Cureus 2024; 16:e72816. [PMID: 39493341 PMCID: PMC11528318 DOI: 10.7759/cureus.72816] [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: 11/01/2024] [Indexed: 11/05/2024] Open
Abstract
Pancreatic cancer is one of the most lethal malignancies, characterized by late diagnosis, rapid progression, and limited treatment options. This literature review comprehensively examines the epidemiology, risk factors, diagnostic challenges, treatment modalities, and prognosis of pancreatic cancer. It highlights the global disparities in incidence and outcomes, exploring the influence of socioeconomic, environmental, and genetic factors on disease progression. In addition, this review discusses recent advancements in diagnostic tools and treatment strategies, identifying gaps in current research and clinical practices. The synthesis aims to inform future research directions and policy-making efforts to reduce the global burden of pancreatic cancer and improve patient outcomes.
Collapse
Affiliation(s)
| | | | - Umar Bin Tariq
- General Surgery, Southmead Hospital Bristol, North Bristol NHS Trust, Bristol, GBR
| | - Arslan Ahmad
- Emergency Medicine, Weston General Hospital, University Hospitals Bristol and Weston NHS Foundation Trust, Weston-super-Mare, GBR
| | - Wajeeha Kiran
- Trauma and Orthopaedics, Morriston Hospital, Swansea, GBR
| | | |
Collapse
|
22
|
Lara P, Quiñonero F, Ortiz R, Prados J, Melguizo C. Nanoparticles Bounded to Interfering RNAs as a Therapy for Pancreatic Cancer: A Systematic Review. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2013. [PMID: 39510122 DOI: 10.1002/wnan.2013] [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: 04/30/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Pancreatic cancer is one of the tumors with poor prognosis and low survival due to late diagnosis, high resistance, and very limited effective therapeutic options. Thus, new pharmacological treatments are necessary to improve the prognosis of patients. In this context, nanoparticles represent an efficient system for transporting and administering therapeutic molecules. Furthermore, siRNA can be used in cancer treatment to selectively inhibit the expression of any target gene. Therefore, nanoparticles associated with siRNA have been tested as a new therapeutic strategy to solve the pancreatic cancer treatment failure in the clinical setting. The current article presents a systematic revision of the literature of the last 10 years in which nanoparticles loading siRNA are used in pancreatic cancer. This research was carried out in three databases (PubMed, Scopus, and Web of Science) obtaining 164 articles from which 37 were selected. Our results show an overall view of the high effectiveness of this new therapy that combines nanoparticles with genetic therapy in pancreatic cancer suggesting that siRNA-based medicines will likely open up a new therapeutic era in the treatment of this type of tumors.
Collapse
Affiliation(s)
- Patricia Lara
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), Granada, Spain
| | - Francisco Quiñonero
- Instituto de Investigación Biosanitaria de Granada, (Ibs.GRANADA), Granada, Spain
| | - Raul Ortiz
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (Ibs.GRANADA), Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (Ibs.GRANADA), Granada, Spain
- Department of Anatomy and Embryology, University of Granada, Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (Ibs.GRANADA), Granada, Spain
- Department of Anatomy and Embryology, University of Granada, Granada, Spain
| |
Collapse
|
23
|
Kelley JK, Kolbeinsson H, Chandana S, Eastburg B, Frisch A, Parker J, Wright GP, Assifi MM, Chung M. Neoadjuvant Chemotherapy and Radiation Improves Recurrence-free and Overall Survival in Resectable and Borderline Resectable Pancreatic Ductal Adenocarcinoma. Am Surg 2024; 90:2762-2768. [PMID: 38676648 DOI: 10.1177/00031348241250043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
OBJECTIVE The objective of this study is to analyze the outcomes of patients with resectable/borderline resectable PDAC who receive total neoadjuvant therapy vs upfront surgery. METHODS AND ANALYSIS Patients who were treated at a single institution from 2006 to 2021 were included. The primary outcome was overall survival (OS). Secondary outcomes included disease free survival (DFS), rates of lymph node positivity, and R0 resection. All survival analyses were performed with intention-to-treat. RESULTS 26 patients received neoadjuvant chemotherapy and radiation (TNT), 28 received neoadjuvant chemotherapy only (NAC), and 168 received upfront surgery. Demographics were comparable across all three groups. Patients who received TNT or NAC had longer OS and DFS compared to the surgery first patients (P < .01). Patients who received TNT had a lymph node positivity rate of 0% at time of surgery compared to 5.3% and 13.3% in the NAC and surgery-first groups, respectively (P < .01). The rate of R0 resection did not differ between groups (P = .17). CONCLUSION Patients with resectable/borderline resectable PDAC who receive neoadjuvant therapy have longer OS and RFS relative to those who receive upfront surgery.
Collapse
Affiliation(s)
- Jesse K Kelley
- Corewell Health West General Surgery, Grand Rapids, MI, USA
| | | | | | - Benjamin Eastburg
- Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Austin Frisch
- Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Jessica Parker
- Corewell Health West Scholarly Activity and Scientific Support, Grand Rapids, MI, USA
| | - G Paul Wright
- Corewell Health West General Surgery, Grand Rapids, MI, USA
- Michigan State University College of Human Medicine, Grand Rapids, MI, USA
- Division of Surgical Oncology, Corewell Health West General Surgery, Grand Rapids, MI, USA
| | - M Mura Assifi
- Corewell Health West General Surgery, Grand Rapids, MI, USA
- Michigan State University College of Human Medicine, Grand Rapids, MI, USA
- Division of Surgical Oncology, Corewell Health West General Surgery, Grand Rapids, MI, USA
| | - Mathew Chung
- Corewell Health West General Surgery, Grand Rapids, MI, USA
- Michigan State University College of Human Medicine, Grand Rapids, MI, USA
- Division of Surgical Oncology, Corewell Health West General Surgery, Grand Rapids, MI, USA
| |
Collapse
|
24
|
Siciliano AC, Forciniti S, Onesto V, Iuele H, Cave DD, Carnevali F, Gigli G, Lonardo E, Del Mercato LL. A 3D Pancreatic Cancer Model with Integrated Optical Sensors for Noninvasive Metabolism Monitoring and Drug Screening. Adv Healthc Mater 2024; 13:e2401138. [PMID: 38978424 DOI: 10.1002/adhm.202401138] [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: 03/26/2024] [Revised: 06/06/2024] [Indexed: 07/10/2024]
Abstract
A distinct feature of pancreatic ductal adenocarcinoma (PDAC) is a prominent tumor microenvironment (TME) with remarkable cellular and spatial heterogeneity that meaningfully impacts disease biology and treatment resistance. The dynamic crosstalk between cancer cells and the dense stromal compartment leads to spatially and temporally heterogeneous metabolic alterations, such as acidic pH that contributes to drug resistance in PDAC. Thus, monitoring the extracellular pH metabolic fluctuations within the TME is crucial to predict and to quantify anticancer drug efficacy. Here, a simple and reliable alginate-based 3D PDAC model embedding ratiometric optical pH sensors and cocultures of tumor (AsPC-1) and stromal cells for simultaneously monitoring metabolic pH variations and quantify drug response is presented. By means of time-lapse confocal laser scanning microscopy (CLSM) coupled with a fully automated computational analysis, the extracellular pH metabolic variations are monitored and quantified over time during drug testing with gemcitabine, folfirinox, and paclitaxel, commonly used in PDAC therapy. In particular, the extracellular acidification is more pronounced after drugs treatment, resulting in increased antitumor effect correlated with apoptotic cell death. These findings highlight the importance of studying the influence of cellular metabolic mechanisms on tumor response to therapy in 3D tumor models, this being crucial for the development of personalized medicine approaches.
Collapse
Affiliation(s)
- Anna Chiara Siciliano
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Stefania Forciniti
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Valentina Onesto
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Helena Iuele
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Donatella Delle Cave
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, National Research Council (Cnr-IGB), Naples, 80131, Italy
| | - Federica Carnevali
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Experimental Medicine, University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Enza Lonardo
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, National Research Council (Cnr-IGB), Naples, 80131, Italy
| | - Loretta L Del Mercato
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| |
Collapse
|
25
|
Karam E, Rondé-Roupie C, Aussilhou B, Hentic O, Rebours V, Lesurtel M, Sauvanet A, Dokmak S. Laparoscopic pancreatoduodenectomy is safe for the treatment of pancreatic ductal adenocarcinoma treated by chemoradiotherapy compared with open pancreatoduodenectomy: A matched case-control study. Surgery 2024:S0039-6060(24)00829-8. [PMID: 39488453 DOI: 10.1016/j.surg.2024.09.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/12/2024] [Accepted: 09/29/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Few studies compared laparoscopic and open pancreatoduodenectomy for pancreatic ductal adenocarcinoma after neoadjuvant chemoradiotherapy. METHODS Retrospective cohort of patients who underwent laparoscopic or open pancreatoduodenectomy for resectable or borderline resectable pancreatic ductal adenocarcinoma after chemoradiotherapy between 2012 and 2023 was analyzed. Open pancreatoduodenectomy patients could theoretically benefit from the laparoscopic approach. We used a 1:2 (laparoscopic-to-open pancreatoduodenectomy) propensity score matching analysis stratified on age, gender, and body mass index. RESULTS We included 128 patients (33 laparoscopic and 95 open pancreatoduodenectomy), and after propensity score matching, 33 laparoscopic pancreatoduodenectomy and 66 open pancreatoduodenectomy were compared. There was no difference in demographic data except for lower tobacco use in laparoscopic pancreatoduodenectomy group (9% vs 30%, P = .023) with similar clinical presentation. Laparoscopic pancreatoduodenectomy compared to open pancreatoduodenectomy showed a longer median operative duration (380 vs 255 minutes, P < .001), shorter median length of resected vein (15 vs 23 mm, P = .01), longer median venous clamping time (29 vs 15 minutes, P = .005), similar median blood loss (300 vs 300 mL, P = .223), similar rate of hard pancreas (97% vs 85%, P = .094), and a larger median size of Wirsung duct (5 vs 4 mm, P = .02). Postoperative outcomes showed similar 90-day mortality rates (3% vs 3%, P > .99), Clavien-Dindo III-IV complications (6% vs 14%, P = .158), median lengths of hospital stay (12 vs 13 days, P = .409), and readmission rates (9% vs 18%, P = .366). Pathologic data showed similar R0 resection rates (88% vs 82%, P = .568). With a similar rate of adjuvant chemotherapy (P = .324) and shorter median follow-up with laparoscopic pancreatoduodenectomy (18 vs 34 months, P = .004), 3-year overall (P = .768) and disease-free (P = .839) survival rates were similar. CONCLUSION In selected patients, laparoscopic pancreatoduodenectomy for pancreatic ductal adenocarcinoma after neoadjuvant chemoradiotherapy appears to be safe and feasible when performed in experienced centers.
Collapse
Affiliation(s)
- Elias Karam
- Department of Hepato-biliary-Pancreatic Surgery and Liver Transplantation, APHP, Hôpital Beaujon, DMU DIGEST, Clichy, France; Visceral Surgery Unit, Tours University Hospital, France
| | - Charlotte Rondé-Roupie
- Department of Hepato-biliary-Pancreatic Surgery and Liver Transplantation, APHP, Hôpital Beaujon, DMU DIGEST, Clichy, France
| | - Béatrice Aussilhou
- Department of Hepato-biliary-Pancreatic Surgery and Liver Transplantation, APHP, Hôpital Beaujon, DMU DIGEST, Clichy, France
| | - Olivia Hentic
- Department of Pancreatology, APHP, Hôpital Beaujon, DMU DIGEST, Clichy, France
| | - Vinciane Rebours
- Department of Pancreatology, APHP, Hôpital Beaujon, DMU DIGEST, Clichy, France; Université de Paris Cité, Paris, France
| | - Mickaël Lesurtel
- Department of Hepato-biliary-Pancreatic Surgery and Liver Transplantation, APHP, Hôpital Beaujon, DMU DIGEST, Clichy, France; Université de Paris Cité, Paris, France
| | - Alain Sauvanet
- Department of Hepato-biliary-Pancreatic Surgery and Liver Transplantation, APHP, Hôpital Beaujon, DMU DIGEST, Clichy, France; Université de Paris Cité, Paris, France
| | - Safi Dokmak
- Department of Hepato-biliary-Pancreatic Surgery and Liver Transplantation, APHP, Hôpital Beaujon, DMU DIGEST, Clichy, France; Université de Paris Cité, Paris, France; Centre de Recherche sur l'Inflammation, INSERM Unité Mixte de Recherche 1149, Clichy, France.
| |
Collapse
|
26
|
Espona-Fiedler M, Patthey C, Lindblad S, Sarró I, Öhlund D. Overcoming therapy resistance in pancreatic cancer: New insights and future directions. Biochem Pharmacol 2024; 229:116492. [PMID: 39153553 DOI: 10.1016/j.bcp.2024.116492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Pancreatic adenocarcinoma (PDAC) is predicted to become the second leading cause of cancer deaths by 2030 and this is mostly due to therapy failure. Limited treatment options and resistance to standard-of-care (SoC) therapies makes PDAC one of the cancer types with poorest prognosis and survival rates [1,2]. Pancreatic tumors are renowned for their poor response to therapeutic interventions including targeted therapies, chemotherapy and radiotherapy. Herein, we review hallmarks of therapy resistance in PDAC and current strategies aiming to tackle escape mechanisms and to re-sensitize cancer cells to therapy. We will further provide insights on recent advances in the field of drug discovery, nanomedicine, and disease models that are setting the ground for future research.
Collapse
Affiliation(s)
- Margarita Espona-Fiedler
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå Universitet, Umeå, Sweden.
| | - Cedric Patthey
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå Universitet, Umeå, Sweden
| | - Stina Lindblad
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden
| | - Irina Sarró
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Universitat de Barcelona, Barcelona, Spain
| | - Daniel Öhlund
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå Universitet, Umeå, Sweden.
| |
Collapse
|
27
|
Zhang B, Zhang B, Wang T, Huang B, Cen L, Wang Z. Integrated bulk and single-cell profiling characterize sphingolipid metabolism in pancreatic cancer. BMC Cancer 2024; 24:1347. [PMID: 39487387 PMCID: PMC11531184 DOI: 10.1186/s12885-024-13114-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Abnormal sphingolipid metabolism (SM) is closely linked to the incidence of cancers. However, the role of SM in pancreatic cancer (PC) remains unclear. This study aims to explore the significance of SM in the prognosis, immune microenvironment, and treatment of PC. METHODS Single-cell and bulk transcriptome data of PC were acquired via TCGA and GEO databases. SM-related genes (SMRGs) were obtained via MSigDB database. Consensus clustering was utilized to construct SM-related molecular subtypes. LASSO and Cox regression were utilized to build SM-related prognostic signature. ESTIMATE and CIBERSORT algorithms were employed to assess the tumour immune microenvironment. OncoPredict package was used to predict drug sensitivity. CCK-8, scratch, and transwell experiments were performed to analyze the function of ANKRD22 in PC cell line PANC-1 and BxPC-3. RESULTS A total of 153 SMRGs were acquired, of which 48 were linked to PC patients' prognosis. Two SM-related subtypes (SMRGcluster A and B) were identified in PC. SMRGcluster A had a poorer outcome and more active SM process compared to SMRGcluster B. Immune analysis revealed that SMRGcluster B had higher immune and stromal scores and CD8 + T cell abundance, while SMRGcluster A had a higher tumour purity score and M0 macrophages and activated dendritic cell abundance. PC with SMRGcluster B was more susceptible to gemcitabine, paclitaxel, and oxaliplatin. Then SM-related prognostic model (including ANLN, ANKRD22, and DKK1) was built, which had a very good predictive performance. Single-cell analysis revealed that in PC microenvironment, macrophages, epithelial cells, and endothelial cells had relatively higher SM activity. ANKRD22, DKK1, and ANLN have relatively higher expression levels in epithelial cells. Cell subpopulations with high expression of ANKRD22, DKK1, and ANLN had more active SM activity. In vitro experiments showed that ANKRD22 knockdown can inhibit the proliferation, migration, and invasion of PC cells. CONCLUSION This study revealed the important significance of SM in PC and identified SM-associated molecular subtypes and prognostic model, which provided novel perspectives on the stratification, prognostic prediction, and precision treatment of PC patients.
Collapse
Affiliation(s)
- Biao Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bolin Zhang
- Department of Visceral, Vascular and Endocrine Surgery, Martin-Luther-University Halle- Wittenberg, University Medical Center Halle, Halle, Germany
| | - Tingxin Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Bingqian Huang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Lijun Cen
- Department of Transfusion Medicine, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China.
- Key Laboratory of Molecular Pathology in Tumors of Guangxi, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China.
| | - Zhizhou Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
28
|
Suto H, Nagao M, Matsukawa H, Fuke T, Ando Y, Oshima M, Takahashi S, Shibata T, Kamada H, Kobara H, Okuyama H, Hirao T, Kumamoto K, Okano K. Relationships between postoperative recurrences and standardized uptake value on 18F-fluorodeoxyglucose-positron emission tomography in patients with resectable and borderline resectable pancreatic ductal adenocarcinoma who underwent curative pancreatic resection after neoadjuvant chemoradiotherapy. Pancreatology 2024; 24:1133-1140. [PMID: 39160121 DOI: 10.1016/j.pan.2024.08.010] [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: 04/14/2024] [Revised: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND This study aimed to examine postoperative recurrence after curative pancreatic resection following neoadjuvant chemoradiotherapy (NACRT) in patients with resectable (R-) and borderline resectable (BR-) pancreatic ductal adenocarcinoma (PDAC), focusing on its relationship with the standardized uptake value (SUV) on 18F-fluorodeoxyglucose-positron emission tomography (FDG-PET). METHOD The postoperative initial recurrence patterns were examined in patients with R- and BR-PDAC who underwent NACRT followed by curative pancreatic resection. Data collected from three prospective clinical trials were retrospectively analysed. RESULTS After a median follow-up of 29 months, 91 (60 %) of 151 patients experienced postoperative recurrence. The median recurrence-free survival (RFS) for all patients was 18 months. The sites of first recurrence were lung-only in 24 (26 %) patients, liver-only in 23 (25 %), local-only in 11 (12 %), peritoneum-only in 10 (11 %), other single site in 5 (5 %), and multiple sites in 19 (21 %) patients. Multivariate analysis identified the maximum standardized uptake value (SUVmax) on FDG-PET at diagnoses ≥5.40 (hazard ratio [HR], 1.62; 95 % confidence interval [CI], 1.01-2.61; p = 0.045) and node-positive pathology (HR, 2.01; 95 % CI, 1.32-3.08; p = 0.001) as significant predictors of RFS. Furthermore, the SUVmax at initial diagnosis and after NACRT correlated with liver metastasis. CONCLUSION R- and BR-PDACs with high SUV on FDG-PET at diagnosis are risk factors for postoperative recurrence. Among patients who undergo surgery after NACRT, those with a high SUVmax at diagnosis or post-NACRT require careful attention for postoperative liver recurrence.
Collapse
Affiliation(s)
- Hironobu Suto
- Department of Gastroenterological Surgery, Faculty of Medicine, Kagawa University, Kagawa, Japan.
| | - Mina Nagao
- Department of Gastroenterological Surgery, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Hiroyuki Matsukawa
- Department of Gastroenterological Surgery, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takuro Fuke
- Department of Gastroenterological Surgery, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yasuhisa Ando
- Department of Gastroenterological Surgery, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Minoru Oshima
- Department of Gastroenterological Surgery, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Shigeo Takahashi
- Department of Radiation Oncology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Toru Shibata
- Department of Radiation Oncology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Hideki Kamada
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Hideki Kobara
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Hiroyuki Okuyama
- Department of Clinical Oncology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Tomohiro Hirao
- Department of Public Health, Kagawa University, Kagawa, Japan
| | - Kensuke Kumamoto
- Department of Gastroenterological Surgery, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Keiichi Okano
- Department of Gastroenterological Surgery, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
29
|
Hua R, Yao HF, Song ZY, Yu F, Che ZY, Gao XF, Huo YM, Liu W, Sun YW, Yang MW, Yang JY, Zhang S, Zhang JF. Evaluation of a new scoring system for assessing nerve invasion in resected pancreatic cancer: A single-center retrospective analysis. Cancer Lett 2024; 603:217213. [PMID: 39244006 DOI: 10.1016/j.canlet.2024.217213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
Nerve invasion (NI) is a characteristic feature of pancreatic cancer. Traditional dichotomous statements on the presence of NI are unreasonable because almost all cases exhibit NI when sufficient pathological sections are examined. The critical implications of NI in pancreatic cancer highlight the need for a more effective criterion. This study included 511 patients, who were categorized into a training group and a testing group at a ratio of 7:3. According to the traditional definition, NI was observed in 91.2 % of patients using five pathological slides in our study. The prevalence of NI increased as more pathological slides were used. The criterion of 'two points of intraneural (endoneural) invasion in the case of four pathological slides' has the highest receiver operating characteristic (ROC) score. Based on this new criterion, NI was proved to be an independent prognostic factor for overall survival (OS) and disease-free survival (DFS) and was also correlated with tumor recurrence (P = 0.004). Interestingly, gemcitabine-based chemotherapy regimen is an independent favorable factor for patients with high NI. In the high NI group, patients who received a gemcitabine-based regimen exhibited a better prognosis than those who did not receive the gemcitabine-based regimen for OS (P = 0.000) and DFS (P = 0.001). In conclusion, this study establishes assessment criteria to evaluate the severity of NI in order to predict patient outcomes.
Collapse
Affiliation(s)
- Rong Hua
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Hong-Fei Yao
- Department of Hepato-Biliary-Pancreatic Surgery, General Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, PR China
| | - Zi-Yu Song
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Feng Yu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Zhao-Yu Che
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Xiao-Fang Gao
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Yan-Miao Huo
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Wei Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Yong-Wei Sun
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Min-Wei Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China.
| | - Jian-Yu Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China.
| | - Shan Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, PR China.
| | - Jun-Feng Zhang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China; Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation, Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, PR China.
| |
Collapse
|
30
|
He H, Zou CF, Jiang YJ, Yang F, Di Y, Li J, Jin C, Fu DL. Recurrence scoring system predicting early recurrence for patients with pancreatic ductal adenocarcinoma undergoing pancreatectomy and portomesenteric vein resection. World J Gastrointest Surg 2024; 16:3185-3201. [PMID: 39575290 PMCID: PMC11577395 DOI: 10.4240/wjgs.v16.i10.3185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/19/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND Pancreatectomy with concomitant portomesenteric vein resection (PVR) enables patients with portomesenteric vein (PV) involvement to achieve radical resection of pancreatic ductal adenocarcinoma, however, early recurrence (ER) is frequently observed. AIM To predict ER and identify patients at high risk of ER for individualized therapy. METHODS Totally 238 patients undergoing pancreatectomy and PVR were retrospectively enrolled and were allocated to the training or validating cohort. Univariate Cox and LASSO regression analyses were performed to construct serum recurrence score (SRS) based on 26 serum-derived parameters. Uni- and multivariate Cox regression analyses of SRS and 18 clinicopathological variables were performed to establish a Nomogram. Receiver operating characteristic curve analysis was used to evaluate the predictive accuracy. Survival analysis was performed using Kaplan-Meier method and log-rank test. RESULTS Independent serum-derived recurrence-relevant factors of LASSO regression model, including postoperative carbohydrate antigen 19-9, postoperative carcinoembryonic antigen, postoperative carbohydrate antigen 125, preoperative albumin (ALB), preoperative platelet to ALB ratio, and postoperative platelets to lymphocytes ratio, were used to construct SRS [area under the curve (AUC): 0.855, 95%CI: 0.786-0.924]. Independent risk factors of recurrence, including SRS [hazard ratio (HR): 1.688, 95%CI: 1.075-2.652], pain (HR: 1.653, 95%CI: 1.052-2.598), perineural invasion (HR: 2.070, 95%CI: 0.827-5.182), and PV invasion (HR: 1.603, 95%CI: 1.063-2.417), were used to establish the recurrence nomogram (AUC: 0.869, 95%CI: 0.803-0.934). Patients with either SRS > 0.53 or recurrence nomogram score > 4.23 were considered at high risk for ER, and had poor long-term outcomes. CONCLUSION The recurrence scoring system unique for pancreatectomy and PVR, will help clinicians in predicting recurrence efficiently and identifying patients at high risk of ER for individualized therapy.
Collapse
Affiliation(s)
- Hang He
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Cai-Feng Zou
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Yong-Jian Jiang
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Feng Yang
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Yang Di
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Ji Li
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Chen Jin
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - De-Liang Fu
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| |
Collapse
|
31
|
Nickerson JL, Cyr C, Arseneau RJ, Lee SN, Condon-Oldreive S, Zogopoulos G, Roberts K, Kim CA, Ng SSW, Haider M, Villalba E, Stephenson L, Tsang E, Johnston B, Gala-Lopez B, Cooper V, Hannon B, Gangloff A, Gill S, Servidio-Italiano F, Ramjeesingh R. Canadian National Pancreas Conference 2023: A Review of Multidisciplinary Engagement in Pancreatic Cancer Care. Curr Oncol 2024; 31:6191-6204. [PMID: 39451765 PMCID: PMC11506161 DOI: 10.3390/curroncol31100461] [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: 08/22/2024] [Revised: 10/02/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Pancreatic cancer is a complex malignancy associated with poor prognosis and high symptom burden. Optimal patient care relies on the integration of various sectors in the healthcare field as well as innovation through research. The Canadian National Pancreas Conference (NPC) was co-organized and hosted by Craig's Cause Pancreatic Cancer Society and The Royal College of Physicians and Surgeons in November 2023 in Montreal, Canada. The conference sought to bridge the gap between Canadian healthcare providers and researchers who share the common goal of improving the prognosis, quality of life, and survival for patients with pancreatic cancer. The accredited event featured discussion topics including diagnosis and screening, value-based and palliative care, pancreatic enzyme replacement therapy, cancer-reducing treatment, and an overview of the current management landscape. The present article reviews the NPC sessions and discusses the presented content with respect to the current literature.
Collapse
Affiliation(s)
- Jessica L. Nickerson
- Allumiqs Corporation, Halifax, NS B3H 0A8, Canada;
- Craig’s Cause Pancreatic Cancer Society, Halifax, NS B3K 5M3, Canada; (C.C.); (R.J.A.); (S.N.L.); (S.C.-O.)
| | - Chloe Cyr
- Craig’s Cause Pancreatic Cancer Society, Halifax, NS B3K 5M3, Canada; (C.C.); (R.J.A.); (S.N.L.); (S.C.-O.)
- Department of Kinesiology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 0A2, Canada
| | - Riley J. Arseneau
- Craig’s Cause Pancreatic Cancer Society, Halifax, NS B3K 5M3, Canada; (C.C.); (R.J.A.); (S.N.L.); (S.C.-O.)
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 0A2, Canada
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Stacey N. Lee
- Craig’s Cause Pancreatic Cancer Society, Halifax, NS B3K 5M3, Canada; (C.C.); (R.J.A.); (S.N.L.); (S.C.-O.)
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 0A2, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Stefanie Condon-Oldreive
- Craig’s Cause Pancreatic Cancer Society, Halifax, NS B3K 5M3, Canada; (C.C.); (R.J.A.); (S.N.L.); (S.C.-O.)
| | | | - Keith Roberts
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK;
| | - Christina A. Kim
- Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada;
| | - Sylvia S. W. Ng
- Section of Medical Oncology and Hematology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0W2, Canada;
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
| | - Masoom Haider
- Joint Department of Medical Imaging, Sinai Health System, Toronto, ON M5G 1X6, Canada;
| | - Eva Villalba
- Quebec Cancer Coalition, Saint-Lambert, QC J4P 2J7, Canada;
| | | | - Erica Tsang
- Department of Medicine, Princess Margaret Cancer Centre, Toronto, ON M5G 2C4, Canada;
| | - Brent Johnston
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Boris Gala-Lopez
- Department of Surgery, Dalhousie University, Halifax, NS B3H 2Y9, Canada;
| | - Valerie Cooper
- South East Local Health Integration Network, Belleville, ON K8N 5K3, Canada;
| | - Breffni Hannon
- Department of Supportive Care, Princess Margaret Cancer Centre, Toronto, ON M5G 2C4, Canada;
| | - Anne Gangloff
- Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada;
| | | | | | - Ravi Ramjeesingh
- Division of Medical Oncology, Dalhousie University, Halifax, NS B3H 2Y9, Canada
| |
Collapse
|
32
|
Groß T, Merboth F, Klimowa A, Kahlert C, Distler M, Weitz J, Welsch T, Müssle B. Impact of an enhanced anti-infection prophylaxis strategy for pancreatoduodenectomy: a single centre analysis. Langenbecks Arch Surg 2024; 409:307. [PMID: 39402424 PMCID: PMC11473572 DOI: 10.1007/s00423-024-03465-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/01/2024] [Indexed: 10/19/2024]
Abstract
INTRODUCTION : Surgical site infection (SSI) after pancreatoduodenectomy (PD) is a significant concern. Targeted antibiotic prophylaxis (pAP) has been tested to mitigate antibiotic resistance patterns, especially after preoperative bile duct stenting. The aim of this study was to investigate the effect of enhanced anti-infective prophylaxis (EAP) on the incidence of superficial and intraabdominal SSI. METHODS All patients who underwent PD at a single centre between May 2018 and May 2021 were retrospectively analysed. A control cohort of patients who received pAP with intravenous cefuroxime and metronidazole and routine intraoperative abdominal lavage according to the surgeons' preferences. Since March 2020, pAP has been changed to piperacillin/tazobactam according to local resistance patterns and combined with routine intraoperative extended abdominal lavage (EIPL). Preoperative selective decontamination of the digestive tract (SDD) has been applied routinely since Jan 2019. RESULTS In total, 163 patients were included. The standard (n = 100) and EAP (n = 63) groups did not significantly differ with regard to pertinent patient and operative characteristics. In the EAP group, the rates of SSI (14% vs. 37%, p = 0.002, total rate: 28%) and urinary tract infection (24% vs. 8%, p = 0.011, total rate 18%) were significantly lower. Other septic complications were not significantly different. In addition, the risk of developing gastrointestinal bleeding and delayed gastric emptying was significantly lower in the EAP group. Multivariate analysis showed that an age > 67 years was a significant risk factor for SSI. CONCLUSION The results indicate that enhanced anti-infective prophylaxis may significantly decrease the incidence of SSI in patients after PD.
Collapse
Affiliation(s)
- Tina Groß
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Ependorf, Hamburg, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Felix Merboth
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
| | - Anna Klimowa
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
| | - Christoph Kahlert
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidberg, Heidelberg, Germany
| | - Marius Distler
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Thilo Welsch
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Ependorf, Hamburg, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Benjamin Müssle
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- Department of General and Visceral Surgery, University Hospital Ulm, Ulm, Germany.
| |
Collapse
|
33
|
Zou X, Ding X, Feng ZK, Ouyang YF, Li HF, Wen K, Wang ZQ, Liu YP, Liu YL, Zhang WJ, Yang Q, Chen SY, Xie YL, Xie RQ, Lin C, Gu CM, Huang PY, Sun R, Hua YJ, You R, Chen MY. A phase 2 trial of gemcitabine plus toripalimab for cisplatin-ineligible patients with recurrent or metastatic nasopharyngeal carcinoma. Cell Rep Med 2024; 5:101779. [PMID: 39413735 PMCID: PMC11514602 DOI: 10.1016/j.xcrm.2024.101779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 04/03/2024] [Accepted: 09/17/2024] [Indexed: 10/18/2024]
Abstract
Cisplatin is a cornerstone chemotherapy for nasopharyngeal carcinoma (NPC); however, certain patients are ineligible for cisplatin-based regimens. This phase 2 trial (NCT04405622) evaluated the efficacy and safety of gemcitabine and toripalimab in previously untreated patients with recurrent or metastatic NPC who were either ineligible for cisplatin or had experienced severe adverse events from prior cisplatin-based treatments. Patients received gemcitabine (1,000 mg/m2) and toripalimab (240 mg) every three weeks for six cycles, followed by toripalimab monotherapy for up to two years. The primary endpoint was the incidence of grade ≥3 adverse events, while secondary endpoints included objective response rate (ORR) and overall survival (OS). Of 30 screened patients, 21 were enrolled. No treatment-related fatalities occurred, with the most frequent adverse events being headache and nausea. The ORR was 61.9%, coupled with a disease control rate of 100%. Overall, gemcitabine plus toripalimab demonstrated low toxicity and promising efficacy for this specific patient cohort.
Collapse
Affiliation(s)
- Xiong Zou
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Xi Ding
- Department of Nasopharyngeal Carcinoma, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, P.R. China
| | - Zheng-Kai Feng
- Department of Nasopharyngeal Carcinoma, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, P.R. China
| | - Yan-Feng Ouyang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Hui-Feng Li
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Kai Wen
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Zhi-Qiang Wang
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, P.R. China
| | - You-Ping Liu
- Department of Nasopharyngeal Carcinoma, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, P.R. China
| | - Yong-Long Liu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Wei-Jing Zhang
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Qi Yang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Si-Yuan Chen
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Yu-Long Xie
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Ruo-Qi Xie
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Chao Lin
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Chen-Mei Gu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Pei-Yu Huang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Rui Sun
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China.
| | - Yi-Jun Hua
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China.
| | - Rui You
- Department of Nasopharyngeal Carcinoma, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, P.R. China; Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China.
| | - Ming-Yuan Chen
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China.
| |
Collapse
|
34
|
Heckler M, Polychronidis G, Kinny-Köster B, Roth S, Hank T, Kaiser J, Michalski C, Loos M. Thrombosis and anticoagulation after portal vein reconstruction during pancreatic surgery: a systematic review. J Gastrointest Surg 2024:101852. [PMID: 39406295 DOI: 10.1016/j.gassur.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/20/2024] [Accepted: 10/05/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Portal vein (PV) resection and reconstruction, which includes the resection and reconstruction of the PV and superior mesenteric vein, enable surgical removal of borderline resectable and locally advanced pancreatic cancer. Thrombosis of the reconstructed PV represents a major cause of early postoperative and long-term morbidity and mortality. No universally accepted standard for anticoagulation exists. This study aimed to assess early and late thrombosis rates after PV reconstruction with special regard to the type of PV reconstruction and anticoagulation regimen and to comprehensively assess thrombotic events and their clinical effect in patients receiving pancreatic surgery with venous resection and reconstruction. METHODS The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed. Studies reporting on PV resection and reconstruction providing data on thrombosis rates were included. The following parameters were assessed: study type, year of publication, number of patients, type/number of PV reconstruction, follow-up period, postoperative mortality, thrombosis rate of the reconstructed PV axis, intraoperative blood loss, and anticoagulation. RESULTS A total of 23 studies with 2751 patients were included in the final analysis. Of note, 670 patients received tangential resection of the PV with venorrhaphy or patch repair, 1505 patients had segmental resection with end-to-end reconstruction, and 576 patients received reconstruction with an interposition graft/conduit. The pooled overall thrombosis rate was 15%. Reconstruction of tangential defects with either venorrhaphy or patch repair and end-to-end repair of segmental defects resulted in a thrombosis rate of 12%. Subgroup analysis according to the type of graft reconstruction revealed the highest occlusion rates of 55% in patients with allogeneic grafts, followed by up to 27% in patients with synthetic PV conduits. Autologous conduits had a thrombosis rate of 10%. Early thrombotic events were detected in 5% of patients after venorrhaphy/patch reconstruction and end-to-end reconstruction. Early events were most common in the allogeneic graft subgroup (22%), followed by synthetic conduits (15%). There were fewer early events in the autologous graft group (7%). Early PV thrombosis was associated with relevant mortality of up to 26%. Anticoagulation regimens varied between studies. CONCLUSION The overall thrombosis rate after PV resection is low. However, among the different reconstruction techniques, allogeneic interposition grafts/conduits had the highest thrombosis rates among the different types of reconstruction after PV resection. No specific anticoagulation strategy can be considered beneficial based on the existing literature.
Collapse
Affiliation(s)
- Max Heckler
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Georgios Polychronidis
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany; Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Benedict Kinny-Köster
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Susanne Roth
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Hank
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Joerg Kaiser
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Christoph Michalski
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Loos
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
35
|
Giani A, Mazzola M, Paterno M, Zironda A, Calcagno P, Zuppi E, De Martini P, Ferrari G. Oncological Outcomes of Open Versus Minimally Invasive Surgery for Ductal Adenocarcinomas of Pancreatic Head: A Propensity Score Matching Analysis. Curr Oncol 2024; 31:6096-6109. [PMID: 39451759 PMCID: PMC11506721 DOI: 10.3390/curroncol31100455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Minimally invasive pancreatic resections (MIPRs) have been shown to be safe and feasible, but there is still a lack of high-level evidence on oncological outcomes for cephalic pancreatic ductal adenocarcinoma (PDAC). The aim of this study was to compare the oncological outcomes of patients undergoing MIPR and open pancreatic resection (OPR) for pancreatic head cancer in a single high-volume center. METHODS Data from a prospectively collected database of patients who underwent radical-intent surgery for resectable and borderline resectable PDAC of the head at our institution between January 2013 and May 2023 were retrieved and analyzed, comparing the surgical and oncological outcomes of MIPR and OPR, using a propensity score matching analysis. RESULTS In the study period, 220 patients were selected. After matching, a total of 81 MIPRs and 81 OPRs were compared. No difference was found regarding R0 rate (OPR 83.9% vs. MIPR 74.1%, p = 0.122). Median overall survival (24 and 31 months for the OPR and MIPR groups, respectively; log rank p = 0.665) and disease-free survival (12 and 21 months for the OPR and MIPR groups, respectively; log rank p = 0.118) did not differ between the groups. The MIPR group was associated with a greater number of harvested lymph nodes (22 vs. 16, p = 0.0008), longer operative time (565 vs. 420 min, p < 0.0001), and shorter length of stay (12 vs. 18 days; p = 0.0001). No differences between the groups were found regarding all other postoperative and pathological outcomes. CONCLUSIONS Regarding oncological outcomes, MIPR appeared to be comparable to OPR for treating patients with PDAC of the head. Despite an increased operative time, MIPR was associated with a greater number of LNs harvested and a shorter length of stay.
Collapse
Affiliation(s)
| | - Michele Mazzola
- Division of Minimally-Invasive Surgical Oncology, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milano, Italy; (A.G.); (M.P.); (A.Z.); (P.C.); (E.Z.); (P.D.M.); (G.F.)
| | | | | | | | | | | | | |
Collapse
|
36
|
Larsson P, Swartling O, Perri G, Vaez K, Holmberg M, Klevebro F, Gilg S, Sparrelid E, Ghorbani P. The impact of chronic obstructive pulmonary disease on risk for complications after pancreatoduodenectomy - a single centre cohort study. HPB (Oxford) 2024:S1365-182X(24)02334-7. [PMID: 39443259 DOI: 10.1016/j.hpb.2024.10.005] [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] [Received: 10/13/2023] [Revised: 05/12/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND The association between chronic obstructive pulmonary disease (COPD) and risk for postoperative complications after pancreatic surgery has not been clarified. The aim of this study was to investigate if COPD is associated with increased risk for postoperative complications after pancreatoduodenectomy. METHODS All patients aged ≥18 years undergoing pancreatoduodenectomy from 2008 to 2019 at a high-volume tertiary centre for pancreatic cancer surgery were included. COPD was defined as an established diagnosis according to the International Statistical Classification of Diseases. The primary outcome was Clavien-Dindo-score (CD)≥ IIIa. RESULTS Out of 1009 available patients, 57 (5.6 %) had a diagnosis of COPD. There was no association between COPD and CD≥ IIIa (25.5 % vs. 29.8 % p-value 0.471). COPD was associated with an increased risk for postoperative pancreatic fistula (POPF) (odds ratio [OR] 3.06, 95 % confidence interval 1.62-5.89; p < 0.001). The 12 months mortality rate was higher among patients with COPD compared to patients without COPD, although not statistically significant (28.07 % vs., 18.17 %, p-value = 0.063). CONCLUSION COPD was associated with increased risk for POPF. These results imply that among patients deemed fit enough to undergo surgery, COPD should be thoroughly evaluated in the risk stratification.
Collapse
Affiliation(s)
- Patrik Larsson
- Department of Upper Abdominal Diseases, Karolinska University Hospital Stockholm, Sweden; Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Oskar Swartling
- Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden; Clinical Epidemiology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Giampaolo Perri
- Department of Upper Abdominal Diseases, Karolinska University Hospital Stockholm, Sweden
| | - Kaveh Vaez
- Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Holmberg
- Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Klevebro
- Department of Upper Abdominal Diseases, Karolinska University Hospital Stockholm, Sweden; Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Stefan Gilg
- Department of Upper Abdominal Diseases, Karolinska University Hospital Stockholm, Sweden; Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ernesto Sparrelid
- Department of Upper Abdominal Diseases, Karolinska University Hospital Stockholm, Sweden; Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Poya Ghorbani
- Department of Upper Abdominal Diseases, Karolinska University Hospital Stockholm, Sweden; Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
37
|
Wang J, Yang J, Narang A, He J, Wolfgang C, Li K, Zheng L. Consensus, debate, and prospective on pancreatic cancer treatments. J Hematol Oncol 2024; 17:92. [PMID: 39390609 PMCID: PMC11468220 DOI: 10.1186/s13045-024-01613-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Pancreatic cancer remains one of the most aggressive solid tumors. As a systemic disease, despite the improvement of multi-modality treatment strategies, the prognosis of pancreatic cancer was not improved dramatically. For resectable or borderline resectable patients, the surgical strategy centered on improving R0 resection rate is consensus; however, the role of neoadjuvant therapy in resectable patients and the optimal neoadjuvant therapy of chemotherapy with or without radiotherapy in borderline resectable patients were debated. Postoperative adjuvant chemotherapy of gemcitabine/capecitabine or mFOLFIRINOX is recommended regardless of the margin status. Chemotherapy as the first-line treatment strategy for advanced or metastatic patients included FOLFIRINOX, gemcitabine/nab-paclitaxel, or NALIRIFOX regimens whereas 5-FU plus liposomal irinotecan was the only standard of care second-line therapy. Immunotherapy is an innovative therapy although anti-PD-1 antibody is currently the only agent approved by for MSI-H, dMMR, or TMB-high solid tumors, which represent a very small subset of pancreatic cancers. Combination strategies to increase the immunogenicity and to overcome the immunosuppressive tumor microenvironment may sensitize pancreatic cancer to immunotherapy. Targeted therapies represented by PARP and KRAS inhibitors are also under investigation, showing benefits in improving progression-free survival and objective response rate. This review discusses the current treatment modalities and highlights innovative therapies for pancreatic cancer.
Collapse
Affiliation(s)
- Junke Wang
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jie Yang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Amol Narang
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jin He
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Christopher Wolfgang
- Department of Surgery, New York University School of Medicine and NYU-Langone Medical Center, New York, NY, USA
| | - Keyu Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA.
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA.
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Multidisciplinary Gastrointestinal Cancer Laboratories Program, the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
38
|
Dong B, Zhang Y, Gao H, Liu J, Li J. Machine Learning Developed a MYC Expression Feature-Based Signature for Predicting Prognosis and Chemoresistance in Pancreatic Adenocarcinoma. Biochem Genet 2024; 62:4191-4214. [PMID: 38245886 DOI: 10.1007/s10528-023-10625-0] [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: 09/03/2023] [Accepted: 12/07/2023] [Indexed: 01/23/2024]
Abstract
MYC has been identified to profoundly influence a wide range of pathologic processes in cancers. However, the prognostic value of MYC-related genes in pancreatic adenocarcinoma (PAAD) remains unclarified. Gene expression data and clinical information of PAAD patients were obtained from The Cancer Genome Atlas (TCGA) database (training set). Validation sets included GSE57495, GSE62452, and ICGC-PACA databases. LASSO regression analysis was used to develop a risk signature for survival prediction. Single-cell sequencing data from GSE154778 and CRA001160 datasets were analyzed. Functional studies were conducted using siRNA targeting RHOF and ITGB6 in PANC-1 cells. High MYC expression was found to be significantly associated with a poor prognosis in patients with PAAD. Additionally, we identified seven genes (ADGRG6, LINC00941, RHOF, SERPINB5, INSYN2B, ITGB6, and DEPDC1) that exhibited a strong correlation with both MYC expression and patient survival. They were then utilized to establish a risk model (MYCsig), which showed robust predictive ability. Furthermore, MYCsig demonstrated a positive correlation with the expression of HLA genes and immune checkpoints, as well as the chemotherapy response of PAAD. RHOF and ITGB6, expressed mainly in malignant cells, were identified as key oncogenes regulating chemosensitivity through EMT. Downregulation of RHOF and ITGB6 reduced cell proliferation and invasion in PANC-1 cells. The developed MYCsig demonstrates its potential in enhancing the management of patients with PAAD by facilitating risk assessment and predicting response to adjuvant chemotherapy. Additionally, our study identifies RHOF and ITGB6 as novel oncogenes linked to EMT and chemoresistance in PAAD.
Collapse
Affiliation(s)
- Biao Dong
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050000, Hebei, China
| | - Yueshan Zhang
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050000, Hebei, China
| | - Han Gao
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050000, Hebei, China
| | - Jia Liu
- Department of Precision Medicine, Accb Biotech. Ltd, Beijing, China
| | - Jiankun Li
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
39
|
Johnson R, McClelland PH, Ahmad SA. Neoadjuvant and Adjuvant Therapy in Resectable Pancreatic Adenocarcinoma. Surg Clin North Am 2024; 104:987-1005. [PMID: 39237173 DOI: 10.1016/j.suc.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
While pancreatic adenocarcinoma requires surgical resection definitive cure, treatment paradigms are shifting toward a neoadjuvant approach to systemic therapy. Rationale is twofold: micro-metastatic disease is likely present in a majority of patients, reinforcing the importance of systemic therapy regardless of resectability; moreover, systemic therapy is well-tolerated and improves surgical outcomes when delivered preoperatively. Second, a neoadjuvant approach allows for selection of biology and patients most likely to benefit from potentially morbid surgery. This review examines the increasing body of evidence in support of empiric neoadjuvant therapy in pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Ryan Johnson
- General Surgery, Division of Surgical Oncology, University of Cincinnati Cancer Institute, University of Cincinnati Medical Center, 231 Albert Sabin Way, ML 0558, Cincinnati, OH 45267, USA
| | - Paul H McClelland
- General Surgery, Division of Surgical Oncology, University of Cincinnati Cancer Institute, University of Cincinnati Medical Center, 231 Albert Sabin Way, ML 0558, Cincinnati, OH 45267, USA
| | - Syed A Ahmad
- Division of Surgical Oncology, University of Cincinnati Cancer Institute, University of Cincinnati Medical Center, 231 Albert Sabin Way, ML 0558, Cincinnati, OH 45267, USA.
| |
Collapse
|
40
|
Leonhardt CS, Gustorff C, Klaiber U, Le Blanc S, Stamm TA, Verbeke CS, Prager GW, Strobel O. Prognostic Factors for Early Recurrence After Resection of Pancreatic Cancer: A Systematic Review and Meta-Analysis. Gastroenterology 2024; 167:977-992. [PMID: 38825047 DOI: 10.1053/j.gastro.2024.05.028] [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: 03/20/2024] [Revised: 05/17/2024] [Accepted: 05/24/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND & AIMS More than half of pancreatic ductal adenocarcinomas (PDACs) recur within 12 months after curative-intent resection. This systematic review and meta-analysis was conducted to identify all reported prognostic factors for early recurrence in resected PDACs. METHODS After a systematic literature search, a meta-analysis was conducted using a random effects model. Separate analyses were performed for adjusted vs unadjusted effect estimates as well as reported odds ratios (ORs) and hazard ratios (HRs). Risk of bias was assessed using the Quality in Prognostic Studies tool, and evidence was rated according to Grading of Recommendations Assessment, Development and Evaluation recommendations. RESULTS After 2903 abstracts were screened, 65 studies were included. Of these, 28 studies (43.1%) defined early recurrence as evidence of recurrence within 6 months, whereas 34 (52.3%) defined it as evidence of recurrence within 12 months after surgery. Other definitions were uncommon. Analysis of unadjusted ORs and HRs revealed 41 and 5 prognostic factors for early recurrence within 6 months, respectively. When exclusively considering adjusted data, we identified 25 and 10 prognostic factors based on OR and HR, respectively. Using a 12-month definition, we identified 38 (OR) and 15 (HR) prognostic factors from unadjusted data and 38 (OR) and 30 (HR) prognostic factors from adjusted data, respectively. On the basis of frequency counts of adjusted data, preoperative carbohydrate antigen 19-9, N status, nondelivery of adjuvant therapy, grading, and tumor size based on imaging were identified as key prognostic factors for early recurrence. CONCLUSIONS Reported prognostic factors of early recurrence vary considerably. Identified key prognostic factors could aid in the development of a risk stratification framework for early recurrence. However, prospective validation is necessary.
Collapse
Affiliation(s)
- Carl-Stephan Leonhardt
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria; Usher Institute, The University of Edinburgh, Edinburgh, United Kingdom.
| | - Charlotte Gustorff
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Ulla Klaiber
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Solange Le Blanc
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Tanja A Stamm
- Institute of Outcomes Research, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Caroline S Verbeke
- Department of Pathology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gerald W Prager
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Oliver Strobel
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
41
|
Funamizu N, Mori S, Sakamoto A, Iwata M, Shine M, Ito C, Uraoka M, Ueno Y, Tamura K, Umeda Y, Aoki T, Takada Y. C-Reactive Protein-to-Albumin Ratio as a Predictive Indicator for Evaluating Tolerability in S-1 Adjuvant Chemotherapy after Curative Surgery for Pancreatic Cancer: An External Validation Cohort Study. Cancers (Basel) 2024; 16:3372. [PMID: 39409992 PMCID: PMC11475895 DOI: 10.3390/cancers16193372] [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: 09/02/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND S-1 in adjuvant chemotherapy (AC) administration after pancreatic cancer (PC) surgery has been standardized in Japan. The Ehime study confirmed that a postoperative higher C-reactive protein-to-albumin ratio (CAR) value predicted the risk of adverse event (AE)-related S-1 non-completion as an AC in patients with PC after curative surgery. This study aimed to investigate the index to predict S-1 tolerance among patients who underwent curative surgery for PC (the Dokkyo study). METHODS This retrospective validation cohort study included 172 patients at the Department of Hepato-Biliary Pancreatic Surgery, Dokkyo Medical University, Japan, from January 2010 to December 2022. All patients underwent nutritional screening using the postoperative CAR. S-1 completion status and its effect on prognosis were systematically followed up and investigated. We conducted a statistical analysis of predictive markers to investigate their association with S-1 completion. RESULTS Patients were categorized into the S-1 completion (N = 91) and non-completion (N = 81) groups. The S-1 completion group demonstrated a significantly lower CAR than the S1 non-completion group. Moreover, the current study revealed a significant difference in the S-1 completion rate, applying the CAR cutoff value of 0.05 established in the Ehime study. Additionally, univariate and multivariate analyses confirmed that a CAR of <0.05 was significantly associated with S-1 completion. CONCLUSIONS The Dokkyo study confirmed the results observed in the Ehime study. Consequently, an increased postoperative CAR value appeared as a universal applicable marker for the risk factor of AE-related S-1 non-completion after curative surgery for patients with PC.
Collapse
Affiliation(s)
- Naotake Funamizu
- Department of Hepato-Biliary Pancreatic and Transplantation Surgery, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon 791-0295, Ehime, Japan; (A.S.); (M.I.); (M.S.); (C.I.); (M.U.); (Y.U.); (K.T.); (Y.U.); (Y.T.)
| | - Shozo Mori
- Department of Hepato-Biliary Pancreatic Surgery, Dokkyo Medical University, Kitakobayashi 880, Mibu, Shimotsugagun 321-0293, Tochigi, Japan; (S.M.); (T.A.)
| | - Akimasa Sakamoto
- Department of Hepato-Biliary Pancreatic and Transplantation Surgery, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon 791-0295, Ehime, Japan; (A.S.); (M.I.); (M.S.); (C.I.); (M.U.); (Y.U.); (K.T.); (Y.U.); (Y.T.)
| | - Miku Iwata
- Department of Hepato-Biliary Pancreatic and Transplantation Surgery, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon 791-0295, Ehime, Japan; (A.S.); (M.I.); (M.S.); (C.I.); (M.U.); (Y.U.); (K.T.); (Y.U.); (Y.T.)
| | - Mikiya Shine
- Department of Hepato-Biliary Pancreatic and Transplantation Surgery, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon 791-0295, Ehime, Japan; (A.S.); (M.I.); (M.S.); (C.I.); (M.U.); (Y.U.); (K.T.); (Y.U.); (Y.T.)
| | - Chihiro Ito
- Department of Hepato-Biliary Pancreatic and Transplantation Surgery, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon 791-0295, Ehime, Japan; (A.S.); (M.I.); (M.S.); (C.I.); (M.U.); (Y.U.); (K.T.); (Y.U.); (Y.T.)
| | - Mio Uraoka
- Department of Hepato-Biliary Pancreatic and Transplantation Surgery, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon 791-0295, Ehime, Japan; (A.S.); (M.I.); (M.S.); (C.I.); (M.U.); (Y.U.); (K.T.); (Y.U.); (Y.T.)
| | - Yoshitomo Ueno
- Department of Hepato-Biliary Pancreatic and Transplantation Surgery, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon 791-0295, Ehime, Japan; (A.S.); (M.I.); (M.S.); (C.I.); (M.U.); (Y.U.); (K.T.); (Y.U.); (Y.T.)
| | - Kei Tamura
- Department of Hepato-Biliary Pancreatic and Transplantation Surgery, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon 791-0295, Ehime, Japan; (A.S.); (M.I.); (M.S.); (C.I.); (M.U.); (Y.U.); (K.T.); (Y.U.); (Y.T.)
| | - Yuzo Umeda
- Department of Hepato-Biliary Pancreatic and Transplantation Surgery, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon 791-0295, Ehime, Japan; (A.S.); (M.I.); (M.S.); (C.I.); (M.U.); (Y.U.); (K.T.); (Y.U.); (Y.T.)
| | - Taku Aoki
- Department of Hepato-Biliary Pancreatic Surgery, Dokkyo Medical University, Kitakobayashi 880, Mibu, Shimotsugagun 321-0293, Tochigi, Japan; (S.M.); (T.A.)
| | - Yasutsugu Takada
- Department of Hepato-Biliary Pancreatic and Transplantation Surgery, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon 791-0295, Ehime, Japan; (A.S.); (M.I.); (M.S.); (C.I.); (M.U.); (Y.U.); (K.T.); (Y.U.); (Y.T.)
| |
Collapse
|
42
|
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; 62:1724-1785. [PMID: 39389105 DOI: 10.1055/a-2338-3716] [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
|
43
|
Yasinzai AQK, Tareen B, Tracy K, Jamil N, Khan M, Ullah H, Raza M, Khan AU, Arif D, Waheed A, Sidhwa F, Misra S, Karki NR, Karim NA, Cavalcante L, Ullah A. Pancreatic ductal adenocarcinoma: exploring clinicopathological trends and racial disparities in a comprehensive U.S. population-based study. Clin Transl Oncol 2024; 26:2618-2628. [PMID: 38615292 DOI: 10.1007/s12094-024-03484-7] [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: 01/25/2024] [Accepted: 03/25/2024] [Indexed: 04/15/2024]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy about 50% of PDAC are metastatic at presentation. In this study, we evaluated PDAC demographics, annual trend analysis, racial disparities, survival rate, and the role of different treatment modalities in localized and metastatic disease. METHODS A total of 144,824 cases of PDAC were obtained from the SEER database from 2000 to 2018. RESULTS The median age was 69 years, with a slightly higher incidence in males (52%) and 80% of all cases were white. Among cases with available data, 43% were grade III tumors and 57% were metastatic. The most common site of metastasis was the liver (15.7%). The annual incidence has increased steadily from 2000 to 2018. The overall observed (OS) 5-year survival rate was 4.4% (95% CI 4.3-4.6%), and 5 years cause-specific survival (CSS) was 5% (95% CI 5.1-5.4%). The 5-year survival with multimodal therapy (chemotherapy, surgery, and radiation) was 22% (95% CI 20.5-22.8%). 5-year CSS for the blacks was lower at 4.7% (95% CI 4.2-5.1%) compared to the whites at 5.3% (95% CI 5.1-5.4%). Multivariate analysis found male gender and black race associated with worse prognosis. Kaplan-Meier survival analysis found multimodal therapy to have the best outcomes in all three stages. CONCLUSION PDAC is an aggressive malignancy with male gender and black race are associated with a poor prognosis. Surgery with chemoradiation was associated with the best overall survival. With steadily increasing rates of PDAC, improved treatment modalities are paramount to improving survival in these patients.
Collapse
Affiliation(s)
| | - Bisma Tareen
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | | | - Nimra Jamil
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Marjan Khan
- Internal Medicine, Marshfield Medical Center, Marshfield, USA
| | - Hafeez Ullah
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Muhammad Raza
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Amin Ullah Khan
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Dauod Arif
- Department of Oncology Developmental Therapeutics, Novant Health, Charlotte, NC, USA
| | - Abdul Waheed
- Department of Surgery, San Joaquin General Hospital, French Camp, CA, 95231, USA
| | - Feroze Sidhwa
- Department of Surgery, San Joaquin General Hospital, French Camp, CA, 95231, USA
| | - Subhasis Misra
- Department of Surgery, BayCare Health System, Tampa, FL, USA
| | - Nabin Raj Karki
- Department of Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, USA
| | - Nagla Abdel Karim
- Department of Hematology-Oncology, Inova Schar Cancer Institute, University of Virginia, Fairfax, VA, 22031, USA
| | - Ludimila Cavalcante
- Department of Oncology Developmental Therapeutics, Novant Health, Charlotte, NC, USA
| | - Asad Ullah
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| |
Collapse
|
44
|
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
|
45
|
Hagerty BL, Fekrmandi F, Schneider T, Fountzilas C, Stiles Z, Kukar M, Calvo B, Cherkassky L. Chemotherapy switch for nonresponse or progression on neoadjuvant chemotherapy for pancreatic adenocarcinoma. J Surg Oncol 2024; 130:1014-1022. [PMID: 39155683 DOI: 10.1002/jso.27803] [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: 07/08/2024] [Accepted: 07/21/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND AND OBJECTIVES Patients with localized pancreatic adenocarcinoma who do not respond to neoadjuvant therapy present a challenge. We sought to define the characteristics and outcomes of those patients to guide clinical practice. METHODS Patients included were those without evidence of biochemical or radiographic response and no evidence of distant progression at the first reassessment after initiation of therapy. RESULTS Of the 45 patients in the cohort, 23 (51.1%) proceeded to surgical exploration with all but one of those undergoing resection. The median overall survival of the study cohort was 28.6 and 48.6 months in those who underwent resection. A total of 13 patients (28.9%) underwent chemotherapy switch (CS) during their course of neoadjuvant therapy. The CS cohort demonstrated higher rates of radiologic progression (25% vs. 10%, p = 0.329), new or worse vascular involvement (58.3% vs. 30%, p = 0.082), and CA 19-9 increase (30.8% vs. 12.9%, p = 0.209) at initial re-staging. Despite this, overall survival was similar between the two groups (20.7 vs. 28.7 months, p = 0.674). CONCLUSION Non-responders to first-line neoadjuvant therapy have poor rates of curative-intent resection. However, resection should be undertaken when feasible. CS may be considered in patients who do not respond to first-line chemotherapy.
Collapse
Affiliation(s)
- Brendan L Hagerty
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Fatemeh Fekrmandi
- Department of Radiation Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Tyce Schneider
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Christos Fountzilas
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Zachary Stiles
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Moshim Kukar
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Benjamin Calvo
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Leonid Cherkassky
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| |
Collapse
|
46
|
Keltner S, Nelson B, Kharofa J. Indications for Radiation in Pancreatic Adenocarcinoma: A Review. Surg Clin North Am 2024; 104:1007-1016. [PMID: 39237160 DOI: 10.1016/j.suc.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Pancreatic adenocarcinoma remains a deadly disease with 5 year overall survival of 10% among all stages. Standard of care for resectable disease remains surgical resection and adjuvant systemic therapy, but paradigms for borderline resectable and unresectable cases remain more nuanced. Radiation has been explored in the neoadjuvant, adjuvant, and definitive settings in a variety of randomized and non-randomized trials with mixed results. There is strong evidence to support the use of neoadjuvant radiation for borderline resectable pancreatic cancer. Utilization of radiation in the adjuvant setting remains unclear while the results of radiation therapy oncology group (RTOG) 0848 are pending.
Collapse
Affiliation(s)
- Samuel Keltner
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Bailey Nelson
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jordan Kharofa
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
47
|
Konishi T, Takano S, Takayashiki T, Suzuki D, Sakai N, Hosokawa I, Mishima T, Nishino H, Suzuki K, Nakada S, Ohtsuka M. Preoperative Prediction of Long-Term Survival After Surgery in Patients with Resectable Pancreatic Ductal Adenocarcinoma. Ann Surg Oncol 2024; 31:6992-7000. [PMID: 38926210 PMCID: PMC11413041 DOI: 10.1245/s10434-024-15648-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Although some clinical trials have demonstrated the benefits of neoadjuvant therapy for resectable pancreatic ductal adenocarcinoma (PDAC), its optimal candidate has not been clarified. This study aimed to detect predictive prognostic factors for resectable PDAC patients who underwent upfront surgery and identify patient cohorts with long-term survival without neoadjuvant therapy. PATIENTS AND METHODS A total of 232 patients with resectable PDAC who underwent upfront surgery between January 2008 and December 2019 were evaluated. RESULTS The median overall survival (OS) time and 5-year OS rate of resectable PDAC with upfront surgery was 31.5 months and 33.3%, respectively. Multivariate analyses identified tumor diameter in computed tomography (CT) ≤ 19 mm [hazard ratio (HR) 0.40, p < 0.001], span-1 within the normal range (HR 0.54, p = 0.023), prognostic nutritional index (PNI) ≥ 44.31 (HR 0.51, p < 0.001), and lymphocyte-to-monocyte ratio (LMR) ≥ 3.79 (HR 0.51, p < 0.001) as prognostic factors that influence favorable prognoses after upfront surgery. According to the prognostic prediction model based on these four factors, patients with four favorable prognostic factors had a better prognosis with a 5-year OS rate of 82.4% compared to others (p < 0.001). These patients had a high R0 resection rate and a low frequency of tumor recurrence after upfront surgery. CONCLUSIONS We identified patients with long-term survival after upfront surgery by prognostic prediction model consisting of tumor diameter in CT, span-1, PNI, and LMR. Evaluation of anatomical, biological, nutritional, and inflammatory factors may be valuable to introduce an optimal treatment strategy for resectable PDAC.
Collapse
Affiliation(s)
- Takanori Konishi
- Department of General Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shigetsugu Takano
- Department of General Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tsukasa Takayashiki
- Department of General Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Daisuke Suzuki
- Department of General Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Nozomu Sakai
- Department of General Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Isamu Hosokawa
- Department of General Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takashi Mishima
- Department of General Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hitoe Nishino
- Department of General Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kensuke Suzuki
- Department of General Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shinichiro Nakada
- Department of General Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Masayuki Ohtsuka
- Department of General Surgery, Chiba University Graduate School of Medicine, Chiba, Japan.
| |
Collapse
|
48
|
Yoo J, Hyun SH, Lee J, Cheon M, Lee KH, Heo JS, Choi JY. Prognostic Significance of 18 F-FDG PET/CT Radiomics in Patients With Resectable Pancreatic Ductal Adenocarcinoma Undergoing Curative Surgery. Clin Nucl Med 2024; 49:909-916. [PMID: 38968550 DOI: 10.1097/rlu.0000000000005363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024]
Abstract
PURPOSE This study aimed to investigate the prognostic significance of PET/CT radiomics to predict overall survival (OS) in patients with resectable pancreatic ductal adenocarcinoma (PDAC). METHODS We enrolled 627 patients with resectable PDAC who underwent preoperative 18 F-FDG PET/CT and subsequent curative surgery. Radiomics analysis of the PET/CT images for the primary tumor was performed using the Chang-Gung Image Texture Analysis toolbox. Radiomics features were subjected to least absolute shrinkage and selection operator (LASSO) regression to select the most valuable imaging features of OS. The prognostic significance was evaluated by Cox proportional hazards regression analysis. Conventional PET parameters and LASSO score were assessed as predictive factors for OS by time-dependent receiver operating characteristic curve analysis. RESULTS During a mean follow-up of 28.8 months, 378 patients (60.3%) died. In the multivariable Cox regression analysis, tumor differentiation, resection margin status, tumor stage, and LASSO score were independent prognostic factors for OS (HR, 1.753, 1.669, 2.655, and 2.946; all P < 0.001, respectively). The time-dependent receiver operating characteristic curve analysis showed that the LASSO score had better predictive performance for OS than conventional PET parameters. CONCLUSIONS The LASSO score using the 18 F-FDG PET/CT radiomics of the primary tumor was the independent prognostic factor for predicting OS in patients with resectable PDAC and may be helpful in determining therapeutic and follow-up plans for these patients.
Collapse
Affiliation(s)
- Jang Yoo
- From the Department of Nuclear Medicine, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju
| | - Seung Hyup Hyun
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Jaeho Lee
- Department of Preventive Medicine, Seoul National University College of Medicine
| | - Miju Cheon
- Department of Nuclear Medicine, Veterans Health Service Medical Center
| | | | - Jin Seok Heo
- Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joon Young Choi
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
| |
Collapse
|
49
|
Oppat KM, Bennett FJ, Maithel SK. A Review of the Indications, Outcomes, and Postoperative Management After Total and Completion Pancreatectomy for Pancreatic Cancer: More Is Not Necessarily Better. Surg Clin North Am 2024; 104:1049-1064. [PMID: 39237163 DOI: 10.1016/j.suc.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
With improvements in surgical technique and advances in pancreatic endocrine and exocrine replacement therapy, the indications for, and threshold to perform, total or completion pancreatectomy in the modern surgical era are ever evolving. The following review will evaluate such indications for pancreatic cancer including pancreatic ductal adenocarcinoma and intraductal papillary mucinous neoplasms. The authors also review the literature on oncologic outcomes of total and completion pancreatectomy for pancreatic cancer. Finally, they discuss the quality of life and postoperative management of the a-pancreatic state.
Collapse
Affiliation(s)
- Kailey M Oppat
- Emory University, 1365B Clifton Road, NE Building B, Suite 4100, Office 4202, Atlanta, GA 30302, USA
| | - Frances J Bennett
- Emory University, 1365B Clifton Road, NE Building B, Suite 4100, Office 4202, Atlanta, GA 30302, USA
| | - Shishir K Maithel
- Emory University, 1365B Clifton Road, NE Building B, Suite 4100, Office 4202, Atlanta, GA 30302, USA.
| |
Collapse
|
50
|
Tazuma S, Sudo T, Ishikawa A, Yamaguchi A, Shibata Y, Ishida Y, Kuraoka K, Uemura K, Takahashi S, Tashiro H. Effects of transmembrane serine protease 4 on the survival in patients with pancreatic ductal adenocarcinoma undergoing surgery followed by adjuvant chemotherapy. Surg Today 2024; 54:1208-1219. [PMID: 38637344 DOI: 10.1007/s00595-024-02824-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/25/2024] [Indexed: 04/20/2024]
Abstract
PURPOSE The transmembrane serine protease 4 (TMPRSS4) gene is upregulated in various human cancers. However, its biological functions in pancreatic ductal adenocarcinoma remain unclear. We examined the expression of TMPRSS4 in pancreatic ductal adenocarcinoma tissues and its correlation with clinicopathological parameters in patients with pancreatic ductal adenocarcinoma who underwent surgery. METHODS The TMPRSS4 expression was immunohistochemically examined in 81 PDAC patients with pancreatic ductal adenocarcinoma. We analyzed the association between the TMPRSS4 expression and clinicopathological factors, the recurrence-free survival (RFS), and the overall survival (OS) and examined the effect of TMPRSS4 expression on cell migration and sensitivity to 5-fluorouracil. RESULTS The expression rate of TMPRSS4 in the samples was 62.9% (51/81). The TMPRSS4 expression was not correlated with any clinicopathological feature. The five-year overall and recurrence-free survival rates were significantly lower in the TMPRSS4-positive group than in the TMPRSS4-negative group. On a multivariate analysis, TMPRSS4 positivity, poorly differentiated histology, and non-adjuvant chemotherapy predicted a poor OS, while TMPRSS4 positivity and poorly differentiated histology predicted a poor RFS. TMPRSS4-silenced pancreatic ductal adenocarcinoma cells showed higher sensitivity to 5- fluorouracil than did the control siRNA-transfected cells. CONCLUSIONS TMPRSS4 can be considered a prognostic factor and therapeutic target for pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Sho Tazuma
- Department of Surgery, Kure Medical Center/Chugoku Cancer Center, National Hospital Organization, 3-1, Kure, Hiroshima, 737-0023, Japan
| | - Takeshi Sudo
- Department of Surgery, Kure Medical Center/Chugoku Cancer Center, National Hospital Organization, 3-1, Kure, Hiroshima, 737-0023, Japan
| | - Akira Ishikawa
- Department of Diagnostic Pathology, Kure Medical Center/Chugoku Cancer Center, National Hospital Organization, 3-1, Kure, Hiroshima, 737-0023, Japan
| | - Atsushi Yamaguchi
- Department of Gastroenterology, Kure Medical Center/Chugoku Cancer Center, National Hospital Organization, 3-1, Kure, Hiroshima, 737-0023, Japan
| | - Yoshiyuki Shibata
- Department of Surgery, Kure Medical Center/Chugoku Cancer Center, National Hospital Organization, 3-1, Kure, Hiroshima, 737-0023, Japan
| | - Yuko Ishida
- Institute for Clinical Laboratory, Kure Medical Center/Chugoku Cancer Center, National Hospital Organization, 3-1, Aoyama, Kure, Hiroshima, 737-0023, Japan
| | - Kazuya Kuraoka
- Department of Diagnostic Pathology, Kure Medical Center/Chugoku Cancer Center, National Hospital Organization, 3-1, Kure, Hiroshima, 737-0023, Japan
| | - Kenichiro Uemura
- Department of Surgery, Graduate School of Biochemical and Health Science, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Shinya Takahashi
- Department of Surgery, Graduate School of Biochemical and Health Science, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Hirotaka Tashiro
- Department of Surgery, Kure Medical Center/Chugoku Cancer Center, National Hospital Organization, 3-1, Kure, Hiroshima, 737-0023, Japan.
| |
Collapse
|