1
|
Westerhoff JM, Scheepens JCM, van Wolffelaar FF, Bernchou U, Bahij R, Erickson BA, Christodouleas JP, Ng SSW, Gani C, Choudhury A, Alongi F, Renz P, Colonias AT, Meijer GJ, Schytte T, Intven MPW, Verkooijen HM, Daamen LA, Hall WA. Quality of Life and Toxicity in Patients With Pancreatic Ductal Adenocarcinoma Treated With Online Adaptive Stereotactic Magnetic Resonance Guided Radiation Therapy. Int J Radiat Oncol Biol Phys 2025; 122:698-708. [PMID: 40180057 DOI: 10.1016/j.ijrobp.2025.03.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 03/19/2025] [Accepted: 03/22/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE Online adaptive magnetic resonance guided radiation therapy (MRgRT) using a hybrid magnetic resonance imaging and linear accelerator enables stereotactic ablative radiation doses to pancreatic tumors. We evaluated patient-reported quality of life (QoL) and clinician-reported toxicity in patients with pancreatic ductal adenocarcinoma after stereotactic MRgRT. METHOD Patients with nonmetastatic pancreatic ductal adenocarcinoma treated with stereotactic MRgRT on a 1.5-Tesla magnetic resonance imaging and linear accelerator according to local standard practices between May 2019 and December 2023 were identified using the international, prospective observational Multi-OutcoMe EvaluatioN of radiation Therapy Using the MR-Linac study (MOMENTUM, NCT04075305). Patient-reported QoL and clinician-reported toxicity were assessed using the European Organization for Research and Treatment of Cancer Core Quality-of-Life Questionnaires and National Cancer Institute Common Terminology Criteria for Adverse Events at baseline, 3, 6, and 12 months of follow-up. Patients with new systemic therapy or resection were censored. Patients with disease progression were additionally censored for a sensitivity analysis. Mean difference (MD) QoL scores from baseline were estimated using a linear mixed model, which were evaluated for clinical relevance (MD ≥ 10) and statistical significance (P ≤ .05). Acute (≤3 months follow-up) and late (3-12 months follow-up) toxicity was captured if grade ≥3. RESULTS A total of 127 patients were included from 8 centers. Treatment dose ranged from 30 to 50 Gy in 5 fractions. Functional QoL domains remained stable over time. A statistically significant and clinically relevant improvement was found for nausea and vomiting (MD -10; 95% CI, -17 to -3; P < .001), and in the sensitivity analysis for nausea and vomiting (MD -11; 95% CI -18 to -3; P < .001) and appetite (MD -14; 95% CI -28 to 0; P = .05), all at 6 months follow-up. No clinically relevant and statistically significant deterioration was found in other domains. New-onset acute and late grade 3 toxicity occurred in 2 patients and 1 patient, respectively. CONCLUSION Stereotactic MRgRT for patients with nonmetastatic pancreatic ductal adenocarcinoma was associated with stable functioning, improved disease-related symptoms, and minimal toxicity up to 12 months after treatment.
Collapse
Affiliation(s)
- Jasmijn M Westerhoff
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jacobien C M Scheepens
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Uffe Bernchou
- Department of Oncology, Odense University Hospital, Odense, Denmark; Laboratory of Radiation Physics, Department of Oncology, Odense University Hospital, Denmark
| | - Rana Bahij
- Department of Oncology, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Beth A Erickson
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - John P Christodouleas
- Elekta AB, Stockholm, Sweden; Department of Radiation Oncology, Hospital of the University of Pennsylvania, Pennsylvania, Philadelphia
| | - Sylvia S W Ng
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre-Odette Cancer Centre, Toronto, Ontario, Canada
| | - Cihan Gani
- Department of Radiation Oncology, University Hospital Tubingen, University of Tubingen, Tubingen, Germany
| | - Ananya Choudhury
- Department of Clinical Oncology, The University of Manchester and The Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Filippo Alongi
- Advanced Radiation Oncology Department, IRCCS Sacro Cuore Don Calabria Hospital, Negrar-Verona, Italy; University of Brescia, Brescia, Italy
| | - Paul Renz
- Radiation Oncology, Allegheny Health Network, Pittsburgh, Pennsylvania
| | | | - Gert J Meijer
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tine Schytte
- Department of Oncology, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Martijn P W Intven
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Helena M Verkooijen
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lois A Daamen
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - William A Hall
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
2
|
Michalet M, Valenzuela G, Nougaret S, Tardieu M, Azria D, Riou O. Development of Multiparametric Prognostic Models for Stereotactic Magnetic Resonance Guided Radiation Therapy of Pancreatic Cancers. Int J Radiat Oncol Biol Phys 2025; 122:678-689. [PMID: 40185208 DOI: 10.1016/j.ijrobp.2025.03.039] [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: 12/20/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/07/2025]
Abstract
PURPOSE Stereotactic magnetic resonance guided adaptive radiation therapy (SMART) is a new option for local treatment of unresectable pancreatic ductal adenocarcinoma, showing interesting survival and local control (LC) results. Despite this, some patients will experience early local and/or metastatic recurrence leading to death. We aimed to develop multiparametric prognostic models for these patients. METHODS AND MATERIALS All patients treated in our institution with SMART for an unresectable pancreatic ductal adenocarcinoma between October 21, 2019, and August 5, 2022 were included. Several initial clinical characteristics as well as dosimetric data of SMART were recorded. Radiomics data from 0.35-T simulation magnetic resonance imaging were extracted. All these data were combined to build prognostic models of overall survival (OS) and LC using machine learning algorithms. RESULTS Eighty-three patients with a median age of 64.9 years were included. A majority of patients had a locally advanced pancreatic cancer (77%). The median OS was 21 months after SMART completion and 27 months after chemotherapy initiation. The 6- and 12-month post-SMART OS was 87.8% (IC95%, 78.2%-93.2%) and 70.9% (IC95%, 58.8%-80.0%), respectively. The best model for OS was the Cox proportional hazard survival analysis using clinical data, with a concordance index inverse probability of censoring weighted of 0.87. Tested on its 12-month OS prediction capacity, this model had good performance (sensitivity 67%, specificity 71%, and area under the curve 0.90). The median LC was not reached. The 6- and 12-month post-SMART LC was 92.4% [IC95%, 83.7%-96.6%] and 76.3% [IC95%, 62.6%-85.5%], respectively. The best model for LC was the component-wise gradient boosting survival analysis using clinical and radiomics data, with a concordance index inverse probability of censoring weighted of 0.80. Tested on its 9-month LC prediction capacity, this model had good performance (sensitivity 50%, specificity 97%, and area under the curve 0.78). CONCLUSIONS Combining clinical and radiomics data in multiparametric prognostic models using machine learning algorithms showed good performance for the prediction of OS and LC. External validation of these models will be needed.
Collapse
Affiliation(s)
- Morgan Michalet
- University Federation of Radiation Oncology of Mediterranean Occitanie, Montpellier Cancer Institute, Montpellier University, Montpellier, France; Montpellier Cancer Research Institute, Montpellier University, Montpellier, France.
| | - Gladis Valenzuela
- Montpellier Cancer Research Institute, Montpellier University, Montpellier, France
| | - Stéphanie Nougaret
- Montpellier Cancer Research Institute, Montpellier University, Montpellier, France
| | - Marion Tardieu
- Montpellier Cancer Research Institute, Montpellier University, Montpellier, France
| | - David Azria
- University Federation of Radiation Oncology of Mediterranean Occitanie, Montpellier Cancer Institute, Montpellier University, Montpellier, France; University Federation of Radiation Oncology of Mediterranean Occitanie, Institut de Cancérologie du Gard, Centre Hospitalier Universitaire Carémeau, Nîmes, France; Montpellier Cancer Research Institute, Montpellier University, Montpellier, France
| | - Olivier Riou
- University Federation of Radiation Oncology of Mediterranean Occitanie, Montpellier Cancer Institute, Montpellier University, Montpellier, France; Montpellier Cancer Research Institute, Montpellier University, Montpellier, France
| |
Collapse
|
3
|
Dekker EN, Janssen QP, van Dam JL, Strijk GJ, Verkolf EMM, Kandala S, Dumas J, Fellah A, O'Reilly EM, Besselink MG, van Eijck CHJ, Homs MYV, van Tienhoven GJ, Wilmink JW, Mustafa DAM, Groot Koerkamp B, Dutch Pancreatic Cancer Group. Blood Sample Collection in Randomized Controlled Trials for Biomarker Discovery and Validation: Experience of the PREOPANC-2 Trial. Ann Surg Oncol 2025; 32:5092-5101. [PMID: 39907876 PMCID: PMC12130148 DOI: 10.1245/s10434-025-16890-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/02/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND This study aimed to investigate the feasibility and yield of blood sample collection in an investigator-initiated nationwide randomized controlled trial (RCT). METHODS In the PREOPANC-2 trial, 375 patients with (borderline) resectable pancreatic cancer were randomly assigned to two neoadjuvant regiments in 19 centers in the Netherlands (2018-2021). Blood sample collection was scheduled at seven time points before, during, and after treatment. The primary outcome was the proportion of successfully collected blood samples at each scheduled time point. RESULTS Of the 375 randomized patients, 12 were excluded from blood sample collection before any treatment. From the remaining 363 patients, 1513 (87 %) of 1748 blood samples were collected, processed, mailed, and centrally stored. The blood samples were collected before treatment from 347 (96 %) of the 363 patients, after the first neoadjuvant cycle from 322 (94 %) of 343 patients, after neoadjuvant treatment (i.e., before surgery) from 260 (83 %) of 313 patients, and after surgery from 210 (77 %) of 271 patients. During the follow-up visits, blood samples were collected from 147 (82 %) of 179 patients 12 months after randomization and from 83 (77 %) of 108 patients after 24 months. A total of 220 samples (13 %) were missing. The most common causes for missing blood samples were scheduling oversights, unsuccessful blood draw attempts, and mailing failures (151 times, 69 %). Blood sample collection was canceled 69 times (31 %) due to COVID-19. CONCLUSION Blood sample collection in the PREOPANC-2 trial had a yield of 96 % before treatment and an overall yield of 87 %. Collection of blood samples for biomarker studies is feasible in a nationwide RCT.
Collapse
Affiliation(s)
- Esther N Dekker
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Quisette P Janssen
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jacob L van Dam
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Gaby J Strijk
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Eva M M Verkolf
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Sridhar Kandala
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jasper Dumas
- Department of Pathology, Tumor Immuno-Pathology Laboratory, Erasmus MC, Rotterdam, The Netherlands
| | - Amine Fellah
- Department of Pathology, Tumor Immuno-Pathology Laboratory, Erasmus MC, Rotterdam, The Netherlands
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marc G Besselink
- Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
| | | | - Marjolein Y V Homs
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Geert-Jan van Tienhoven
- Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
| | - Johanna W Wilmink
- Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
| | - Dana A M Mustafa
- Department of Pathology, Tumor Immuno-Pathology Laboratory, Erasmus MC, Rotterdam, The Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| | | |
Collapse
Collaborators
L V Beerepoot, M L van Bekkum, B A Bonsing, H Bos, K P Bosscha, S A Bouwense, L Brouwer-Hol, A M E Bruynzeel, O R Busch, G Cirkel, P P L O Coene, J W B de Groot, B C M Haberkorn, I H J T de Hingh, T M Karsten, G Kazemier, M B van der Kolk, M S L Liem, O J L Loosveld, S A C Luelmo, C M Luyer, J S D Mieog, V B Nieuwenhuijs, J J M E Nuyttens, D Ten Oever, G A Patijn, H C van Santvoort, M W J Stommel, M M Streppel, A Ten Tije, E Versteijne, J de Vos-Geelen, R F de Wilde,
Collapse
|
4
|
Abuelafia AM, Santofimia-Castaño P, Estaras M, Grasso D, Chuluyan E, Lomberk G, Urrutia R, Dusetti N, Fraunhoffer N, Iovanna J. KRAS inhibition reverses chemotherapy resistance promoted by therapy-induced senescence-like in pancreatic ductal adenocarcinoma. Transl Oncol 2025; 57:102421. [PMID: 40382842 PMCID: PMC12143771 DOI: 10.1016/j.tranon.2025.102421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 05/01/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Emerging evidence suggests that chemotherapy can accumulate senescent-like cells within tumor tissues, a phenomenon linked to therapy resistance. The aim of this study is to analyze the senescence-like state of after-treatment persistent cells associated with KRAS mutational status to offer a therapeutic strategy to target these cells in pancreatic ductal adenocarcinoma (PDAC). EXPERIMENTAL DESIGN Three commercial cell lines and five patient-derived primary cell cultures with different KRAS statuses were studied following gemcitabine treatment. Senescence-like status was assessed using SA-β-gal, together with cell cycle regulators such as p21. Additionally, KRAS mutations were modulated using MRTX1133 and AMG-510, and the signaling pathways ERK and AKT were analyzed and modulated in vitro. Finally, p21 expression, associated with the senescence-like state, on patient outcomes and treatment response was analyzed in publicly available bulk RNA-seq and single-nucleus datasets. RESULTS We observed an overexpression of p21 alongside an increase in SA-β-gal signal in response to gemcitabine treatment, indicating the induction of a senescence-like state. Specific inhibition of KRAS G12D or G12C mutations reduced SA-β-gal signal and sensitized PDAC cells to gemcitabine. Moreover, ERK inhibition but not AKT inhibition decreased SA-β-gal signal. Additionally, we characterized p21 expression levels in relation to patient outcomes and found that they are modulated by treatment. CONCLUSIONS This dual-targeted therapeutic strategy holds promises for overcoming the challenges posed by KRAS-driven cancers, particularly in addressing the formidable obstacle of pancreatic cancer.
Collapse
Affiliation(s)
- Analia Meilerman Abuelafia
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, Equipe labélisée Ligue Nationale contre le cancer, France; Programa franco-argentino de estudio del Cáncer de Páncreas, Argentina
| | - Patricia Santofimia-Castaño
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, Equipe labélisée Ligue Nationale contre le cancer, France; Programa franco-argentino de estudio del Cáncer de Páncreas, Argentina
| | - Matias Estaras
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, Equipe labélisée Ligue Nationale contre le cancer, France
| | - Daniel Grasso
- Programa franco-argentino de estudio del Cáncer de Páncreas, Argentina; Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina; Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Eduardo Chuluyan
- Programa franco-argentino de estudio del Cáncer de Páncreas, Argentina; Buenos Aires University, Center for Pharmacological and Botanical Studies, Faculty of Medicine, National Council for Scientific and Technical Research, Buenos Aires C1121ABG, Argentina; Buenos Aires University, Faculty of Medicine, Department of Microbiology, Parasitology and Immunology, Buenos Aires C1121ABG, Argentina
| | - Gwen Lomberk
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Raul Urrutia
- Genomic Science and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, Equipe labélisée Ligue Nationale contre le cancer, France; Programa franco-argentino de estudio del Cáncer de Páncreas, Argentina
| | - Nicolas Fraunhoffer
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, Equipe labélisée Ligue Nationale contre le cancer, France; Programa franco-argentino de estudio del Cáncer de Páncreas, Argentina; Buenos Aires University, Center for Pharmacological and Botanical Studies, Faculty of Medicine, National Council for Scientific and Technical Research, Buenos Aires C1121ABG, Argentina.
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, Equipe labélisée Ligue Nationale contre le cancer, France; Programa franco-argentino de estudio del Cáncer de Páncreas, Argentina; Hospital de Alta Complejidad El Cruce, Florencio Varela, Buenos Aires, Argentina; University Arturo Jauretche, Florencio Varela, Buenos Aires, Argentina.
| |
Collapse
|
5
|
Daniel N, Farinella R, Belluomini F, Fajkic A, Rizzato C, Souček P, Campa D, Hughes DJ. The relationship of the microbiome, associated metabolites and the gut barrier with pancreatic cancer. Semin Cancer Biol 2025; 112:43-57. [PMID: 40154652 DOI: 10.1016/j.semcancer.2025.03.002] [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] [Received: 11/30/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Pancreatic cancers have high mortality and rising incidence rates which may be related to unhealthy western-type dietary and lifestyle patterns as well as increasing body weights and obesity rates. Recent data also suggest a role for the gut microbiome in the development of pancreatic cancer. Here, we review the experimental and observational evidence for the roles of the oral, gut and intratumoural microbiomes, impaired gut barrier function and exposure to inflammatory compounds as well as metabolic dysfunction as contributors to pancreatic disease with a focus on pancreatic ductal adenocarcinoma (PDAC) initiation and progression. We also highlight some emerging gut microbiome editing techniques currently being investigated in the context of pancreatic disease. Notably, while the gut microbiome is significantly altered in PDAC and its precursor diseases, its utility as a diagnostic and prognostic tool is hindered by a lack of reproducibility and the potential for reverse causality in case-control cohorts. Future research should emphasise longitudinal and mechanistic studies as well as integrating lifestyle exposure and multi-omics data to unravel complex host-microbiome interactions. This will allow for deeper aetiologic and mechanistic insights that can inform treatments and guide public health recommendations.
Collapse
Affiliation(s)
- Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | | | | | - Almir Fajkic
- Department of Pathophysiology Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | | | - Pavel Souček
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic; Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland.
| |
Collapse
|
6
|
Chen G, Li T, Duan R, Liang W, Li B, Xie X, Yang L, Shuai X, Meng X. Cognate Nanovaccine Promotes Tertiary Lymphoid Structures Function and Strengthens Immune Cell Cross-Talk by Targeting Exhausted T Cells in Nonimmunogenic Cancers. ACS NANO 2025; 19:21385-21399. [PMID: 40478679 DOI: 10.1021/acsnano.5c01280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2025]
Abstract
Tertiary lymphoid structures (TLSs) serve as hubs for immune cell activation and coordination to generate qualitative local immune responses within tumors. However, the effect of TLSs in pancreatic adenocarcinoma is limited by a poorly immunogenic tumor microenvironment and severe T-cell exhaustion. In this study, we found that tumor-infiltrating T cells, particularly TLS-associated T cells, predominantly exhibit terminal exhaustion characterized by high T-cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) expression in patients with pancreatic cancer, affecting their contribution to antitumor immunity. Thus, we developed a therapeutic nanovaccine by fusing antigen-sensitized dendritic cell membranes with TIM-3-targeted lipid nanoparticles (MLP-aTIM-3) to synergistically reverse T-cell exhaustion. The prepared nanovaccine provides cognate antigens and costimulation to the exhausted T cells by the TIM-3/aTIM-3 interaction. In vitro and in vivo studies demonstrate that targeting T-cell exhaustion through the MLP-aTIM-3 not only restores T-cell reactivity but also promotes the formation and maturation of TLSs, leading to superior antitumor efficacy in an orthotopic pancreatic cancer model. Additionally, the therapeutic efficacy of MLP-aTIM-3 extends to other tumor models, such as liver metastasis and colorectal cancer. Our study suggests that targeting T-cell exhaustion while enhancing TLS function with MLP-aTIM-3 offers a promising strategy for improving the immune response in nonimmunogenic cancers.
Collapse
Affiliation(s)
- Gengjia Chen
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Tan Li
- Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Rui Duan
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Weiye Liang
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Bo Li
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaoxue Xie
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Long Yang
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaochun Meng
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| |
Collapse
|
7
|
Ponz de Leon Pisani R, Altieri G, Stasio RC, Lazzano P, Reni M, Falconi M, Vanella G, Arcidiacono PG, Capurso G. Gastrointestinal symptoms in the journey of pancreatic cancer patients. Expert Rev Anticancer Ther 2025. [PMID: 40489240 DOI: 10.1080/14737140.2025.2517888] [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: 04/08/2025] [Revised: 06/04/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy frequently arising with nonspecific and overlooked gastrointestinal symptoms. Gastroenterologists are typically the first specialists to encounter these patients, positioning them to play a pivotal role not only in early diagnosis, but also in the ongoing management of the disease's complex symptom burden. AREAS COVERED This review explored gastrointestinal symptoms in patients with PDAC (ranging from pain and diarrhea to anorexia, jaundice, and nausea) and outlined both tumor- and treatment-related causes. A literature review based on non-systematic PubMed search updated to April 2025 was conducted to summarize current diagnostic strategies, medical, endoscopic therapies, and multidisciplinary management approaches. In addition, we present original data from a single-center cohort, suggesting that the involvement of gastroenterologists leads to more comprehensive management of gastrointestinal symptom control and supportive care. EXPERT OPINION Collaboration among specialists is essential for optimizing patient outcomes in the multidisciplinary management of PDAC. Gastroenterologists' 'stewardship' significantly contributes to prompt diagnosis, symptom control, quality of life preservation, and prognosis. Future priorities should focus on strengthening integration within care pathways, fostering interdisciplinary coordination, and implementing shared clinical tools to enhance comprehensive patient care. A well-structured team-based approach is key to advancing holistic PDAC management.
Collapse
Affiliation(s)
- Ruggero Ponz de Leon Pisani
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gabriele Altieri
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rosa Claudia Stasio
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pilar Lazzano
- Gastroenterology and Hepatology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Michele Reni
- Oncology Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- "Vita-Salute" San Raffaele University, Milan, Italy
| | - Massimo Falconi
- "Vita-Salute" San Raffaele University, Milan, Italy
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppe Vanella
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Giorgio Arcidiacono
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- "Vita-Salute" San Raffaele University, Milan, Italy
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- "Vita-Salute" San Raffaele University, Milan, Italy
| |
Collapse
|
8
|
Li X, Chen Y, Qiao G, Ni J, Chen T, Wang Y, Wu C, Zhang Q, Ma T, Gao S, Zhang M, Shen Y, Wu J, Yu J, Que R, Zhang X, Sun K, Xiao W, Jiang T, Bai X, Liang T. 5-Year survival rate over 20 % in pancreatic ductal adenocarcinoma: A retrospective study from a Chinese high-volume center. Cancer Lett 2025; 619:217658. [PMID: 40118244 DOI: 10.1016/j.canlet.2025.217658] [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: 02/28/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
Standardized clinical management of pancreatic adenocarcinoma (PDAC) remains challenging and high-volume centers provide essential insights for establishing effective multimodal treatment approaches. This retrospective observational study evaluated the impact of standardized, multimodal clinical management on survival outcomes in patients with PDAC across all stages, based on NCCN guidelines resectability criteria, at a high-volume center. From 2019, 4143 patients were diagnosed with PDAC, with 3268 patients receiving further treatment, including surgical resection and/or systemic therapy. The median overall survival (OS) was 18.5 (95 %CI 17.5-19.4) months for the treated cohort and the 5-year survival rate reached 23.3 %. Patients who underwent surgical resection had significantly improved median OS compared to those who received non-surgical treatments (28.4 months vs. 13.0 months, P < 0.001), with corresponding 5-year survival rates of 31.6 % vs. 15.0 %. Moreover, the patients who received NAT followed by surgical resection had improved survival outcomes compared to those who underwent upfront surgical resection in both resectable (median OS: 37.5 months vs. 28.9 months, P < 0.01) and borderline resectable group (median OS: 31.8 months vs. 18.4 months, P < 0.001). This study demonstrated a 5-year survival rate exceeding 20 % for PDAC across all stages at our center. The application of evidence-based treatment strategies through the multidisciplinary team, accompanying with standardized and comprehensive therapeutic managements, high patient adherence, have been considered as critical determinants in enhancing therapeutic efficacy and improving long-term prognosis for patients with PDAC.
Collapse
Affiliation(s)
- Xiang Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Yiwen Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Guoliang Qiao
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Ni
- Information Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Chen
- Department of Radiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yangyang Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Chengyi Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Tao Ma
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Shunliang Gao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Min Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Yan Shen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Jian Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Jun Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Risheng Que
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Xiaochen Zhang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke Sun
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenbo Xiao
- Department of Radiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian'an Jiang
- Department of Ultrasound, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
9
|
Dreyer SB, Beer P, Hingorani SR, Biankin AV. Improving outcomes of patients with pancreatic cancer. Nat Rev Clin Oncol 2025; 22:439-456. [PMID: 40329051 DOI: 10.1038/s41571-025-01019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 05/08/2025]
Abstract
Research studies aimed at improving the outcomes of patients with pancreatic ductal adenocarcinoma (PDAC) have brought about limited progress, and in clinical practice, the optimized use of surgery, chemotherapy and supportive care have led to modest improvements in survival that have probably reached a plateau. As a result, PDAC is expected to be the second leading cause of cancer-related death in Western societies within a decade. The development of therapeutic advances in PDAC has been challenging owing to a lack of actionable molecular targets, a typically immunosuppressive microenvironment, and a disease course characterized by rapid progression and clinical deterioration. Yet, the progress in our understanding of PDAC and identification of novel therapeutic opportunities over the past few years is leading to a strong sense of optimism in the field. In this Perspective, we address the aforementioned challenges, including biological aspects of PDAC that make this malignancy particularly difficult to treat. We explore specific areas with potential for therapeutic advances, including targeting mutant KRAS, novel strategies to harness the antitumour immune response and approaches to early detection, and propose mechanisms to improve clinical trial design and to overcome various community and institutional barriers to progress.
Collapse
Affiliation(s)
- Stephan B Dreyer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- Department of Hepatobiliary Surgery, Royal Liverpool University Hospital, Liverpool, UK
| | - Philip Beer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- Hull York Medical School, University of York, York, UK
| | - Sunil R Hingorani
- Department of Internal Medicine, Division of Hemotology/Oncology, University of Nebraska Medical Center, Omaha, NE, USA
- Pancreatic Cancer Center of Excellence, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK.
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK.
| |
Collapse
|
10
|
Chuong MD, Ashman J, Jethwa K, Kharofa J, Kim H, Koay E, Ludmir E, Miller E, Nelson B, Reyngold M, Sanford N, Chang D. Moving From the Background Toward the Spotlight: A Critical Review of Radiation Therapy for Locally Advanced Pancreas Cancer. Int J Radiat Oncol Biol Phys 2025; 122:294-312. [PMID: 40032056 DOI: 10.1016/j.ijrobp.2025.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/31/2025] [Accepted: 02/16/2025] [Indexed: 03/05/2025]
Abstract
Radiation therapy (RT) for locally advanced pancreatic cancer (LAPC) continues to be controversial. Advances in both systemic therapy and RT techniques have changed the landscape of LAPC management in recent years. Clinical outcomes of ablative RT have been encouraging, and randomized clinical trials may clarify the role of RT for LAPC. We present a contemporary critical review of key aspects regarding optimal patient selection, radiation dose escalation techniques, novel radiosensitizers and radioprotectors, and treatment response assessment for LAPC.
Collapse
Affiliation(s)
- Michael D Chuong
- Department of Radiation Oncology, Miami Cancer Institute, Miami, Florida.
| | - Jonathan Ashman
- Department of Radiation Oncology, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Krishan Jethwa
- Department of Radiation Oncology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Jordan Kharofa
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Hyun Kim
- Department of Radiation Oncology, Washington University in St. Louis, Missouri.
| | - Eugene Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ethan Ludmir
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eric Miller
- Department of Radiation Oncology, Ohio State University, Columbus, Ohio
| | - Bailey Nelson
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Marsha Reyngold
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nina Sanford
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Daniel Chang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
11
|
Liu Y, Ding J, Li S, Jiang A, Chen Z, Quan M. LPA released from dying cancer cells after chemotherapy inactivates Hippo signaling and promotes pancreatic cancer cell repopulation. Cell Oncol (Dordr) 2025; 48:655-671. [PMID: 39903418 PMCID: PMC12119658 DOI: 10.1007/s13402-025-01038-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2025] [Indexed: 02/06/2025] Open
Abstract
PURPOSE The Hippo pathway in the tumorigenesis and progression of PDAC, with lysophosphatidic acid (LPA) regulating the Hippo pathway to facilitate cancer progression. However, the impact of the Hippo signaling pathway on tumor repopulation in PDAC remains unreported. METHODS Direct and indirect co-culture models to investigate gemcitabine-induced apoptotic cells can facilitate the repopulation of residual tumor cells. Mass spectrometry analysis was conducted to assess the impact of gemcitabine treatment on the lipid metabolism of pancreatic cancer cells. ELISA assays confirmed gemcitabine promotes the release of LPA from apoptotic pancreatic cancer cells. The expression of Yes-associated protein 1 (YAP1) elucidated the underlying mechanism by which dying cells induce tumor repopulation using qRT-PCR and Western blot. We studied the biological function of pancreatic cancer cells using CCK-8, colony formation, and transwell invasion assays in vitro. Co-culture models were used to validate the impact of Hippo pathway on tumor repopulation, while flow cytometry was employed to assess the sensitivity of pancreatic cancer cells to gemcitabine in the context of Hippo pathway. RESULTS Gemcitabine-induced dying cells released LPA in a dose-dependent manner, which promoted the proliferation, clonal formation, and invasion of pancreatic cancer cells. Mechanistic studies showed that gemcitabine and LPA facilitated the translocation of YAP1 and induced the inactivation of the Hippo pathway. YAP1 overexpression significantly enhanced the activity of autotaxin, leading to stimulated pancreatic cancer cells to secrete LPA. This mechanism orchestrated a self-sustaining LPA-Hippo feedback loop, which drove the repopulation of residual tumor cells. Simultaneously, it was observed that suppressing LPA and YAP1 expression enhanced the sensitivity of pancreatic cancer cells to gemcitabine. CONCLUSION Our investigation indicated that targeting the LPA-YAP1 signaling pathway could serve as a promising strategy to augment the overall therapeutic efficacy against PDAC.
Collapse
Affiliation(s)
- Yuzhi Liu
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Jie Ding
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Shumin Li
- Department of Oncology and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Anyi Jiang
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Zhiqin Chen
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China.
| | - Ming Quan
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China.
| |
Collapse
|
12
|
Brada LJH, Schouten TJ, Daamen LA, Seelen LWF, Walma MS, van Dam R, de Hingh IH, Liem MSL, de Meijer VE, Patijn GA, Festen S, Stommel MWJ, Bosscha K, Besselink MG, van Santvoort HC, Molenaar IQ. Evaluation of Short and Long-term Outcomes After Resection in Patients With Locally Advanced Versus (Borderline) Resectable Pancreatic Cancer. Ann Surg 2025; 281:1026-1031. [PMID: 38557955 DOI: 10.1097/sla.0000000000006289] [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: 04/04/2024]
Abstract
OBJECTIVE To evaluate short and long-term outcomes after pancreatectomy in patients with locally advanced pancreatic cancer (LAPC) compared with patients with (borderline) resectable pancreatic cancer [(B)RPC]. BACKGROUND Selected patients diagnosed with LAPC are increasingly undergoing resection after induction chemotherapy. To evaluate the benefit of this treatment approach, it is helpful to compare outcomes in resected patients with primary LAPC to outcomes in resected patients with primary (B)RPC. METHODS Two prospectively maintained nationwide databases were used for this study. Patients with (B)RPC undergoing upfront tumor resection and patients with resected LAPC after induction therapy were included. Outcomes were postoperative pancreas-specific complications, 90-day mortality, pathologic outcomes, disease-free interval, and overall survival. RESULTS Overall, 879 patients were included; 103 with LAPC (12%) and 776 with (B)RPC (88%). Patients with LAPC had a lower World Health Organization performance score and ageadjusted Charlson Comorbidity Index. Postoperative pancreas-specific complications were comparable between groups, except delayed gastric emptying grade C, which occurred more often in patients with LAPC (9% vs 3%, P = 0.03). Ninety-day mortality was comparable. About half of the patients in both groups [54% in LAPC vs 48% in (B)RPC, P = 0.21] had a radical resection (R0). Disease-free interval was 13 months in both groups ( P = 0.12) and overall survival from the date of diagnosis was 24 months in patients with LAPC and 19 months in patients with (B)RPC ( P = 0.34). CONCLUSIONS In our nationwide prospective databases, pancreas-specific complications, mortality, and survival in patients with LAPC after pancreatectomy are comparable with those undergoing resection for (B)RPC. These outcomes suggest that postoperative morbidity and mortality after tumor resection in carefully selected patients with LAPC are acceptable.
Collapse
Affiliation(s)
- Lilly J H Brada
- Department of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Utrecht, The Netherlands
- Department of Surgery, Amsterdam UMC, Location University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Thijs J Schouten
- Department of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Utrecht, The Netherlands
| | - Lois A Daamen
- Department of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Utrecht, The Netherlands
| | - Leonard W F Seelen
- Department of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Utrecht, The Netherlands
| | - Marieke S Walma
- Department of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Utrecht, The Netherlands
| | - Ronald van Dam
- Department of Surgery, Maastricht UMC+, Maastricht, The Netherlands
- Department of General and Viseral Surgery, University Hospital Aachen, Aachen, Germany
| | - Ignace H de Hingh
- Department of Epidemiology, GROW-School for Oncology and Developmental Biology, Maastricht University, Aachen, Maastricht, The Netherlands
- Department of Surgery, Catharina Hospital, Eindhoven, The Netherlands
| | - Mike S L Liem
- Department of Surgery, Medical Spectrum Twente, Enschede, The Netherlands
| | - Vincent E de Meijer
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Gijs A Patijn
- Department of Surgery, Isala, Zwolle, The Netherlands
| | | | - Martijn W J Stommel
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Koop Bosscha
- Department of Surgery, Jeroen Bosch Hospital, 's-Hertogenbosch, The Netherlands
| | - Marc G Besselink
- Department of Surgery, Amsterdam UMC, Location University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Hjalmar C van Santvoort
- Department of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Utrecht, The Netherlands
| | - Izaak Quintus Molenaar
- Department of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
13
|
Fu Y, Chen L, Lv N, Wang J, Yu S, Fang Q, Xin W. miR-135b-5p/PDE3B Axis Regulates Gemcitabine Resistance in Pancreatic Cancer Through Epithelial-Mesenchymal Transition. Mol Carcinog 2025; 64:1119-1130. [PMID: 40170518 DOI: 10.1002/mc.23914] [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: 11/19/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/03/2025]
Abstract
Gemcitabine-based chemotherapy is an effective treatment for pancreatic cancer (PC), but gemcitabine resistance frequently compromises the therapeutic efficacy, resulting in clinical chemotherapeutic failure and a poor prognosis for patients. In this study, we investigated the mechanisms of gemcitabine chemoresistance in PC by examining the roles of microRNAs linked to gemcitabine resistance and their downstream signaling pathways. In vitro experiments were performed to alter miR-135b-5p levels in PC parental and drug-resistant cells to probe its function. miR-135b-5p targets PDE3B was confirmed by using RNA-seq technology to screen for gemcitabine-resistance-associated mRNAs in PC. A series of rescue experiments were performed after cotransfection, demonstrating that PDE3B could reverse miR-135b-5p-mediated chemoresistance and epithelial-mesenchymal transition (EMT). These findings indicate that the miR-135b-5p/PDE3B axis generates resistance by stimulating the EMT signaling pathway, which provides new insights into gemcitabine chemoresistance in PC.
Collapse
Affiliation(s)
- Yuxuan Fu
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Liangsheng Chen
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Neng Lv
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Jia Wang
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Shuwei Yu
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Qilu Fang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Wenxiu Xin
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Zhang J, Kong X, Zhou B, Li R, Yu Z, Zhu J, Xi Q, Li Y, Zhao Z, Zhang R. Metabolic reprogramming of drug resistance in pancreatic cancer: mechanisms and effects. Mol Aspects Med 2025; 103:101368. [PMID: 40398192 DOI: 10.1016/j.mam.2025.101368] [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: 01/19/2025] [Revised: 05/10/2025] [Accepted: 05/10/2025] [Indexed: 05/23/2025]
Abstract
Pancreatic cancer is a highly aggressive gastrointestinal malignancy, often termed the "king of cancers" due to its notoriously high mortality rate. Its clinical characteristics, including late diagnosis, low surgical resectability, high recurrence rates, significant chemoresistance, and poor prognosis have collectively driven the persistent rise in incidence and mortality. Despite ongoing advancements in therapeutic strategies, the management of pancreatic cancer, particularly at advanced stages, remains challenging. Chemotherapy remains the mainstay of current treatment. However, the prevalent problem of chemotherapy resistance poses a significant obstacle to effective treatment. Metabolic reprogramming, characterized by alterations in glucose metabolism, lipid biosynthesis, and amino acid utilization, supports the high energy demands and rapid proliferation of cancer cells. Emerging evidence suggests that these metabolic changes, possibly mediated by epigenetic mechanisms, also contribute to tumorigenesis and metastasis. These findings highlight the critical role of metabolic alterations in pancreatic cancer pathogenesis. This review explores the relationship between metabolic reprogramming and chemotherapy resistance, discussing underlying mechanisms and summarizing preclinical studies and drug development targeting metabolism. The aim is to provide a comprehensive perspective on potential therapeutic strategies for pancreatic cancer.
Collapse
Affiliation(s)
- Jinyi Zhang
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Xueqing Kong
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Boyan Zhou
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Rui Li
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhaoan Yu
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Jinrong Zhu
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Qing Xi
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yan Li
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zichao Zhao
- Department of Emergency Medicine, Shaodong People's Hospital, Shaodong City, Hunan Province, China.
| | - Rongxin Zhang
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
15
|
Chan AHY, Zhao Y, Tan HL, Chua DW, Ng KYY, Lee SY, Lee JJX, Tai D, Goh BKP, Koh YX. Clinical Outcomes of Neoadjuvant Therapy Versus Upfront Surgery in Resectable Pancreatic Cancer: Systematic Review and Meta-analysis of Latest Randomized Controlled Trials. Ann Surg Oncol 2025; 32:4094-4107. [PMID: 39987384 DOI: 10.1245/s10434-024-16674-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: 07/23/2024] [Accepted: 11/23/2024] [Indexed: 02/24/2025]
Abstract
BACKGROUND Survival and surgical benefits of neoadjuvant treatments (NAT) in resectable pancreatic cancer (RPC) remains unclear. The role of NAT in providing additional benefits to reduce biological aggressiveness and recurrence is worth elucidating. We assessed the latest randomized controlled trials (RCTs). METHODS A systematic review and meta-analysis was performed including trials published from inception to February 2024 to evaluate survival, surgical, and short-term oncological benefits with RCTs for RPC, comparing NAT with upfront surgery. RESULTS Eight RCTs with 982 patients were analyzed. RPC treated with NAT conferred better median disease-free survival (DFS) compared to upfront surgery (HR = 0.66, p = 0.01) with a significantly improved R0 resection (RR = 1.20, p = 0.04) and pN0 rate (RR = 1.68, p < 0.001). These benefits did not translate into overall survival benefits (HR = 0.81, p = 0.06). Postoperative major morbidity and mortality did not differ significantly between treatment approaches. No significant difference was noted in resection rate (RR = 0.95, p = 0.21). However, a significantly lower surgical exploration rate was exhibited in the NAT group (RR = 0.84, p = 0.007). CONCLUSION NAT conferred better DFS with significantly improved R0 resection rate and pN0 rate compared with upfront surgery. Our findings highlight the potential benefits of NAT in enhancing survival, surgical, and short-term oncological outcomes without increasing postoperative risks.
Collapse
Affiliation(s)
- Anna Ho Yin Chan
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore, Singapore
- Duke-National University of Singapore Graduate Medical School, Singapore, Singapore
| | - Yun Zhao
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore, Singapore
| | - Hwee Leong Tan
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore, Singapore
| | - Darren Weiquan Chua
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore, Singapore
| | - Kennedy Yao Yi Ng
- Duke-National University of Singapore Graduate Medical School, Singapore, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Suat Ying Lee
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Joycelyn Jie Xin Lee
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - David Tai
- Duke-National University of Singapore Graduate Medical School, Singapore, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Brian Kim Poh Goh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore, Singapore
- Duke-National University of Singapore Graduate Medical School, Singapore, Singapore
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Ye Xin Koh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore, Singapore.
- Duke-National University of Singapore Graduate Medical School, Singapore, Singapore.
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore.
| |
Collapse
|
16
|
Hüttner FJ, Klotz R, Giese NA, Kong B, Ahmed A, Merz D, Pöchmann A, Burghaus I, Hackert T, Strobel O, Mihaljevic AL, Michalski CW, Büchler MW, Diener MK. Pancreatic resection with perioperative drug repurposing of propranolol and etodolac - the phase II randomized controlled PROSPER trial. Langenbecks Arch Surg 2025; 410:168. [PMID: 40402347 PMCID: PMC12098435 DOI: 10.1007/s00423-025-03735-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2025] [Accepted: 05/07/2025] [Indexed: 05/23/2025]
Abstract
PURPOSE The perioperative period is characterized by psychological stress and inflammatory reactions that can contribute to disease recurrence or metastatic spread. These reactions are mediated particularly by catecholamines and prostaglandins. The PROSPER trial aimed to evaluate whether a perioperative drug repurposing with a non-selective betablocker (propranolol) and a COX-2 inhibitor (etodolac) is feasible and safe in the setting of pancreatic cancer surgery. METHODS Patients undergoing partial pancreatoduodenectomy for pancreatic cancer were randomized to perioperative treatment with propranolol and etodolac or placebo. Main safety endpoint was the rate of serious adverse events (SAE) and the main feasibility endpoint was adherence. Overall and disease-free survival (DFS) as well as recurrences were assessed as efficacy parameters and the trial was accompanied by a translational study. RESULTS The trial was prematurely closed due to slow recruitment. 26 patients were randomized, but 6 never started trial medication. Finally, 9 patients received the trial medication and 11 patients placebo. There were 6 SAE in the treatment vs. 14 in the placebo group. Adherence was lower in the treatment group, but without statistically significance. Median DFS was 16.36 months (95%-CI 1.18 - not reached) in verum vs. 11.25 (95%-CI 2.2 - 17.25) in placebo group. The rate of distant recurrences was 11.1% in verum vs. 54.5% in placebo group. CONCLUSION There were no safety concerns, but the trial intervention was not feasible given slow recruitment and limited adherence. However, the translational study and preliminary efficacy data revealed some promising findings, warranting further investigation. REGISTRATION DRKS00014054.
Collapse
Affiliation(s)
- Felix J Hüttner
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Department of General, Visceral and Thoracic Surgery, Klinikum Nürnberg, Prof.-Ernst-Nathan-Str. 1, 90419, Nuremberg, Germany
| | - Rosa Klotz
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Study Center of the German Surgical Society (SDGC), Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Nathalia A Giese
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Bo Kong
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Azaz Ahmed
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Department of Cancer Immunology and Cancer Immunotherapy, German Cancer Research Center, Heidelberg, Germany
| | - Daniela Merz
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Alexandra Pöchmann
- Department of Cancer Immunology and Cancer Immunotherapy, German Cancer Research Center, Heidelberg, Germany
- Helmholtz Institute for Translational Oncology (HI-TRON), Mainz, Germany
| | - Ina Burghaus
- Coordination Centre for Clinical Trials (KKS), Medical Faculty & Heidelberg University Hospital, Heidelberg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver Strobel
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - André L Mihaljevic
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Department of General, Visceral and Transplantation Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Christoph W Michalski
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Markus W Büchler
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Botton-Champalimaud Pancreatic Cancer, Champalimaud Foundation, Lisbon, Portugal
| | - Markus K Diener
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany.
- Department of General, Visceral and Thoracic Surgery, Klinikum Nürnberg, Prof.-Ernst-Nathan-Str. 1, 90419, Nuremberg, Germany.
| |
Collapse
|
17
|
Wei F, Li W, Zhou T, Feng L, Yuan X, Zhang L. FAM111B and FANCD2, a dual expression signature, defines a distinct phenotype of pancreatic cancer. Cancer Cell Int 2025; 25:185. [PMID: 40405284 PMCID: PMC12101032 DOI: 10.1186/s12935-025-03819-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 05/08/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Despite various treatment strategies, the incidence and mortality of pancreatic cancer (PC) are among the highest for malignant tumors. Furthermore, there is a lack of effective molecular typing and targeted therapy to treat PC subtypes. METHODS Multiplex immunofluorescence experiments were performed to explore the roles of FAM111B, FANCD2, KRAS and TP53 in human PC tissues. Kaplan-Meier survival curves were generated and a nomogram was prepared for prognostic prediction. Protein correlations were analyzed using human PC tissues and TCGA and GEO data. Pathways analysis, immunoanalysis, and drug susceptibility analysis were performed based on information in the TCGA database. RESULTS Our results indicate that expression of FAM111B and FANCD2 is correlated in human PC tissues and comprises a dual expression signature with predictive value for the prognosis of PC. Using information in public databases, we confirmed the oncogenic relevance of FAM111B and FANCD2 in PC and identified a positive correlation between FAM111B, FANCD2, TP53 and KRAS.FAM111B and FANCD2 jointly regulate ferroptosis, mitotic nuclear division, and nuclear division pathways. Both proteins were demonstrated to be positively correlated with markers of CD4 + Th2 cells and PD-L1 in the tumor microenvironment. Furthermore, drug sensitivity analysis suggested that patients with high FAM111B or FANCD2 expression were highly sensitive to chemotherapeutic and targeted drugs, indicating that these proteins may serve as predictors of treatment efficacy. CONCLUSION Elevated dual expression of FAM111B and FANCD2 is indicative of poor prognosis, alters the immune microenvironment, and exhibits sensitivity to certain therapeutic agents. Consequently, the high FAM111B/FANCD2 expression subtype may represent a novel and distinct phenotype of PC.
Collapse
Affiliation(s)
- Fang Wei
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Wanying Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Ting Zhou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Lijuan Feng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Lihong Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
18
|
Rincon-Torroella J, Dal Molin M, Mog B, Han G, Watson E, Wyhs N, Ishiyama S, Ahmedna T, Minn I, Azad N, Bettegowda C, Papadopoulos N, Kinzler KW, Zhou S, Vogelstein B, Gabrielson K, Sur S. ME3BP-7 is a targeted cytotoxic agent that rapidly kills pancreatic cancer cells expressing high levels of monocarboxylate transporter MCT1. eLife 2025; 13:RP94488. [PMID: 40391649 PMCID: PMC12092006 DOI: 10.7554/elife.94488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025] Open
Abstract
Nearly 30% of pancreatic ductal adenocarcinomas (PDACs) exhibit a marked overexpression of monocarboxylate transporter 1 (MCT1) offering a unique opportunity for therapy. However, biochemical inhibitors of MCT1 have proven unsuccessful in clinical trials. In this study, we present an alternative approach using 3-bromopyruvate (3BP) to target MCT1 overexpressing PDACs. 3BP is a cytotoxic agent that is known to be transported into cells via MCT1, but its clinical usefulness has been hampered by difficulties in delivering the drug systemically. We describe here a novel microencapsulated formulation of 3BP (ME3BP-7), which is effective against a variety of PDAC cells in vitro and remains stable in serum. Furthermore, systemically administered ME3BP-7 significantly reduces pancreatic cancer growth and metastatic spread in multiple orthotopic models of pancreatic cancer with manageable toxicity. ME3BP-7 is, therefore, a prototype of a promising new drug, in which the targeting moiety and the cytotoxic moiety are both contained within the same single small molecule.
Collapse
Affiliation(s)
- Jordina Rincon-Torroella
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neurosurgery, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Marco Dal Molin
- Department of Surgery, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Brian Mog
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Gyuri Han
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Evangeline Watson
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Nicolas Wyhs
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Shun Ishiyama
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins UniversityBaltimoreUnited States
| | - Taha Ahmedna
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins UniversityBaltimoreUnited States
| | - Il Minn
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Nilofer Azad
- Department of Oncology, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Chetan Bettegowda
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neurosurgery, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer CenterBaltimoreUnited States
| | - Nickolas Papadopoulos
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer CenterBaltimoreUnited States
| | - Kenneth W Kinzler
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer CenterBaltimoreUnited States
| | - Shibin Zhou
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer CenterBaltimoreUnited States
| | - Bert Vogelstein
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer CenterBaltimoreUnited States
| | - Kathleen Gabrielson
- Department of Oncology, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins UniversityBaltimoreUnited States
| | - Surojit Sur
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer CenterBaltimoreUnited States
| |
Collapse
|
19
|
Shu QA, Huang XD, Wu J, Liang XR, Wang YL, Yu JM, Wang ZW, Liu N, Wang JH, Xie N. ARHGAP11A is a potential prognostic biomarker and therapeutic target for pancreatic adenocarcinoma. Int J Biochem Cell Biol 2025; 185:106804. [PMID: 40398712 DOI: 10.1016/j.biocel.2025.106804] [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: 02/27/2024] [Revised: 06/23/2024] [Accepted: 05/16/2025] [Indexed: 05/23/2025]
Abstract
The prognosis of pancreatic adenocarcinoma remains dismal up to now, partly owing to a lack of clinically feasible therapeutic targets. ARHGAP11A, a member of Rho GTPase-activating proteins family, has been reported as a prognostic biomarker and oncogene for multiple cancers. However, the specific effects of ARHGAP11A in pancreatic adenocarcinoma remain obscure. In this study, we explored and validated the oncogenic role of ARHGAP11A in pancreatic adenocarcinoma via thorough bioinformatics analyses of public databases and our own RNA-seq data, and in vitro experiments. We found that ARHGAP11A was significantly upregulated in pancreatic adenocarcinoma and correlated with poor clinical outcomes of pancreatic adenocarcinoma patients. Functional enrichment analyses revealed that ARHGAP11A-related genes were enriched in pathways of cell cycle and cell apoptosis. Further molecular functional experiments on ARHGAP11A knockdown pancreatic adenocarcinoma cell lines demonstrated that ARHGAP11A knockdown significantly inhibited cell proliferation, promoted cell cycle arrest in the G1/S stage, and induced cell apoptosis. In comparison, overexpression of ARHGAP11A could remarkably promote the proliferation, cell cycle progression, and apoptosis resistance of pancreatic adenocarcinoma cells. In conclusion, our study demonstrates that ARHGAP11A plays an oncogenic role in pancreatic adenocarcinoma, thus providing a novel therapeutic target and prognostic biomarker for patients with pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Qiu-Ai Shu
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xin-di Huang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Jian Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xi-Ru Liang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Ya-Lan Wang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Jin-Min Yu
- Division of Gastroenterology, Xi'an Central Hospital, Xi'an, Shaanxi 710003, China
| | - Zi-Wei Wang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Na Liu
- Department of Gastroenterology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, Hainan 570100, China.
| | - Jin-Hai Wang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Ning Xie
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| |
Collapse
|
20
|
Zhu Y, Sun R, Fan J, Ma H, Sun B. CSF1-CAR Specifically Targets CSF1R+ Pancreatic Cancer Cells and Tumor-Associated Macrophages. J Immunother 2025:00002371-990000000-00141. [PMID: 40375821 DOI: 10.1097/cji.0000000000000563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/18/2025] [Indexed: 05/18/2025]
Abstract
SUMMARY A highly suppressive tumor immune microenvironment and nonspecific target endow malignant tumors with CAR-T cells. CSF1R is highly expressed on pancreatic cancer tissues compares with normal tissues in GEPIA database and M2 macrophages mainly contributing to the suppressive tumor microenvironment (TME), suggesting that CSF1R is a suitable antigen. CSF1 is the natural ligand of CSF1R, so we constructed a CSF1-CAR and tested its cytotoxic effect on tumor cells and macrophages in vitro. Our results demonstrated that CSF1-CAR-T cells can lyse tumor cells dependent on CSF1R expression. Meanwhile, CSF1-CAR-T also lyse CSF1R+ M2 macrophages, suggesting that CSF1-CAR-T cells play a role in eliminating tumor cells and remodeling the TME.
Collapse
Affiliation(s)
- Yongjie Zhu
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Cell Engineering and Immunotherapy, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruipu Sun
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Cell Engineering and Immunotherapy, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiawei Fan
- General (specialized) Department, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital& Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Haiyan Ma
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Cell Engineering and Immunotherapy, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi province, P.R. China
| | - Bin Sun
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Cell Engineering and Immunotherapy, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
21
|
Ducreux M, Desgrippes R, Rinaldi Y, Di Fiore F, Guimbaud R, Evesque L, Bachet JB, Vanelslander P, Lecomte T, Capitain O, Parzy A, Bolliet M, Etienne PL, Forestier J, El Hajbi F, Bignon AL, Lebrun-Ly V, De Sousa Carvalho N, Texier M, Bouche O. PRODIGE 29-UCGI 26 (NEOPAN): A Phase III Randomized Trial Comparing Chemotherapy With FOLFIRINOX or Gemcitabine in Locally Advanced Pancreatic Carcinoma. J Clin Oncol 2025:JCO2402210. [PMID: 40378359 DOI: 10.1200/jco-24-02210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 03/11/2025] [Accepted: 04/03/2025] [Indexed: 05/18/2025] Open
Abstract
PURPOSE More than 30% of patients with pancreatic cancer are unresectable because of the local extension with a median overall survival (OS) of <1 year. Combination of fluorouracil (FU), oxaliplatin, and irinotecan (FOLFIRINOX) is superior to gemcitabine in the treatment of metastatic pancreatic cancer, but standard of care remains gemcitabine in locally advanced pancreatic cancer (LAPC). METHODS Patients with histologically proven LAPC not suitable for surgery, Eastern Cooperative Oncology Group WHO performance status (PS) ≤1 were eligible. Random assignment was stratified by center, tumor localization (pancreas head yes/no), WHO PS (0 v 1), and age (≤60 years v >60 years). Patients received FOLFIRINOX or gemcitabine for 6 months. The primary end point was progression-free survival (PFS). Main secondary end points were OS, time to treatment failure, quality of life, and safety. One hundred seventy patients (142 events) were needed to detect an increase of 3 months in PFS with 80% power (log-rank test, 5% two-sided α). RESULTS One hundred seventy one patients age 35-84 years were included and followed for a maximum of 5 years. With a median follow-up of 59.6 months (95% CI, 42.3 to not reached), 168 events were observed and the median PFS was 9.7 months (95% CI, 7.0 to 11.7) with FOLFIRINOX versus 7.7 months (95% CI, 6.2 to 9.2) with gemcitabine, hazard ratio (HR), 0.7 (95% CI, 0.5 to 1.0), P = .04. The median OS was 15.7 months (95% CI, 11.9 to 20.4) in the FOLFIRINOX group versus 15.4 months (95% CI, 11.7 to 18.6) in the gemcitabine group, HR, 1.02 (95% CI, 0.73 to 1.43), P = .95. CONCLUSION Results confirm that FOLFIRINOX improves PFS significantly compared with gemcitabine and is well tolerated in LAPC. No significant difference in OS was observed between both groups.
Collapse
Affiliation(s)
- Michel Ducreux
- Gustave Roussy Cancer Center, Tumor Cells Dynamics, INSERM U1279, Université Paris-Saclay, Villejuif, France
| | - Romain Desgrippes
- Hepato-Gastroenterology and Digestive Oncology Department, Centre Hospitalier de Saint Malo, Saint Malo, France
| | | | - Frédéric Di Fiore
- Gastroenterology, CHU Hôpitaux de Rouen-Charles Nicolle, Rouen, France
| | - Rosine Guimbaud
- Digestive Oncology, Centre Hospitalier Rangueil, Toulouse, France
| | | | - Jean-Baptiste Bachet
- Sorbonne University, Hepato-Gastroenterology Department, Groupe Hospitalier Pitié Salpetrière, APHP, Paris, France
| | | | - Thierry Lecomte
- Hepatogastroenterology and Digestive Oncology Departement, Hôpital Trousseau, Chambray-Les-Tours, France
| | - Olivier Capitain
- Medical Oncology, Institut de Cancérologie de l'Ouest-Centre Paul Papin, Angers, France
| | - Aurélie Parzy
- Digestive Pathology, Centre Francois Baclesse, Caen, France
| | - Marion Bolliet
- Gastroenterology, Hôpitaux Civils de Colmar, Colmar, France
| | - Pierre-Luc Etienne
- Medical Oncology, Hôpital Privé des Côtes d'Armor, Plerin sur Mer, France
| | | | - Farid El Hajbi
- Urology and Digestive Oncology, Centre Oscar Lambret, Lille, France
| | | | | | | | - Matthieu Texier
- Biostatistics, Gustave Roussy Institut de Cancérologie, Villejuif, France
| | | |
Collapse
|
22
|
Roscigno G, Jacobs S, Toledo B, Borea R, Russo G, Pepe F, Serrano MJ, Calabrò V, Troncone G, Giovannoni R, Giovannetti E, Malapelle U. The potential application of stroma modulation in targeting tumor cells: focus on pancreatic cancer and breast cancer models. Semin Cancer Biol 2025:S1044-579X(25)00060-4. [PMID: 40373890 DOI: 10.1016/j.semcancer.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer development and spreading being considered as "the dark side of the tumor". Within this term tumor cells, immune components, supporting cells, extracellular matrix and a myriad of bioactive molecules that synergistically promote tumor development and therapeutic resistance, are included. Recent findings revealed the profound impacts of TME on cancer development, serving as physical support, critical mediator and biodynamic matrix in cancer evolution, immune modulation, and treatment outcomes. TME targeting strategies built on vasculature, immune checkpoints, and immuno-cell therapies, have paved the way for revolutionary clinical interventions. On this basis, the relevance of pre-clinical and clinical investigations has rapidly become fundamental for implementing novel therapeutical strategies breaking cell-cell and cell -mediators' interactions between TME components and tumor cells. This review summarizes the key players in the breast and pancreatic TME, elucidating the intricate interactions among cancer cells and their essential role for cancer progression and therapeutic resistance. Different tumors such breast and pancreatic cancer have both different and similar stroma features, that might affect therapeutic strategies. Therefore, this review aims to comprehensively evaluate recent findings for refining breast and pancreatic cancer therapies and improve patient prognoses by exploiting the TME's complexity in the next future.
Collapse
Affiliation(s)
- Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy.
| | - Sacha Jacobs
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| | - Belen Toledo
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain.
| | - Roberto Borea
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| | - Gianluca Russo
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Maria Jose Serrano
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy; GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception Group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain.
| | - Viola Calabrò
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Roberto Giovannoni
- Department of Biology, Genetic Unit, University of Pisa, Via Derna 1, 56126 Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy.
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| |
Collapse
|
23
|
Abreu de Carvalho LF, Gryspeerdt F, Ceelen W, Geboes K, Ribeiro S, Hoorens A, Vandenbussche N, Claes KBM, Lecluyse C, Anisau A, Van Ongeval J, Lybaert W, Triest L, Vervaecke A, Sas S, Claerhout B, Beyls C, Sie M, Berrevoet F. Prediction of surgical resectability after FOLFIRINOX chemotherapy for borderline resectable and locally advanced pancreatic cancer (PeRFormanCe): a multicenter prospective trial - trial protocol. BMC Surg 2025; 25:204. [PMID: 40361171 PMCID: PMC12070518 DOI: 10.1186/s12893-025-02938-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy is used in borderline resectable (BR) and locally advanced (LA) pancreatic ductal adenocarcinoma (PDAC) to increase resection rate and improve cancer outcome. However, there is a need for better prediction of resectability. The aim of this prospective, single arm study is to improve prediction of surgical resection by using radiomics and liquid biopsy. METHODS In this multicentric trial, 45 patients with BR or LA PADC will undergo neoadjuvant chemotherapy with FOLFIRINOX. An intention to treat analysis will be performed. The primary endpoint is the accuracy of the prediction of surgical resection. Secondary endpoints are overall survival and disease-free survival from the date of diagnosis, R0 and R1 resection rates, histopathological response, postoperative complications, patient reported outcomes with quality of life and health economic analysis. Translational research with multi-omics and radiomics based on computed tomography and magnetic resonance imaging aims to identify factors predictive of surgical resectability and survival. The primary hypothesis is that these strategies can increase the accuracy of predicting surgical resection. DISCUSSION Improved prediction of resectability is necessary in BR and LA PDAC. We aim to investigate whether a combination of clinical, radiological, and multi-omics profiling in liquid biopsies can successfully predict resectability and thus optimize the therapeutic decision tree. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05298722. Date of registration: March 28, 2022.
Collapse
Affiliation(s)
- Luís Filipe Abreu de Carvalho
- Department of HPB surgery and liver transplantation, Ghent University Hospital, Corneel Heymanslaan 10, Ghent, 9000, Belgium.
| | - Filip Gryspeerdt
- Department of HPB surgery and liver transplantation, Ghent University Hospital, Corneel Heymanslaan 10, Ghent, 9000, Belgium
| | - Wim Ceelen
- Department of Gastrointestinal surgery, Ghent University Hospital, Ghent, Belgium
| | - Karen Geboes
- Department of Gastroenterology, Division of Digestive Oncology, Ghent University Hospital, Ghent, Belgium
| | - Suzane Ribeiro
- Department of Gastroenterology, Division of Endoscopy and Digestive Oncology, Ghent University Hospital, Ghent, Belgium
| | - Anne Hoorens
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Nele Vandenbussche
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of biomolecular medicine, Ghent University, Ghent, Belgium
- CRIG (Cancer Research Institute Ghent), Ghent, Belgium
| | - Kathleen B M Claes
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of biomolecular medicine, Ghent University, Ghent, Belgium
- CRIG (Cancer Research Institute Ghent), Ghent, Belgium
| | - Clarisse Lecluyse
- Department of Medical Imaging, Ghent University Hospital, Ghent, Belgium
| | - Aliaksandr Anisau
- Department of Medical Imaging, Ghent University Hospital, Ghent, Belgium
| | - Johan Van Ongeval
- Department of Gastroenterology, Division of Digestive Oncology, Ghent Sint-Lucas hospital, Ghent, Belgium
| | - Willem Lybaert
- Department of Gastroenterology, Division of Digestive Oncology, VITAZ hospital, Sint-Niklaas, Belgium
| | - Lars Triest
- Department of Gastroenterology, Division of Digestive Oncology, Sint-Blasius hospital, Dendermonde, Belgium
| | - Andrew Vervaecke
- Department of Gastroenterology, Zeno Hospital, Knokke-Heist and Blankenberge, Knokke-Heist, Belgium
| | - Steven Sas
- Department of Gastroenterology, Division of Digestive Oncology, Jan Palfijn hospital, Ghent, Belgium
| | - Barbara Claerhout
- Department of Gastroenterology, Division of Digestive Oncology, Eeklo Hospital AZ ALMA, Eeklo, Belgium
| | - Carolien Beyls
- Department of Gastroenterology, Division of Digestive Oncology, Oudenaarde hospital, Oudenaarde, Belgium
| | - Mark Sie
- Department of Medical Oncology, Division of Digestive Oncology, Zorgsaam hospital, Terneuzen, The Netherlands
| | - Frederik Berrevoet
- Department of HPB surgery and liver transplantation, Ghent University Hospital, Corneel Heymanslaan 10, Ghent, 9000, Belgium
| |
Collapse
|
24
|
Qian G, Zhang H, Liu Y, Shribak M, Eliceiri KW, Provenzano PP. Computationally Enabled Polychromatic Polarized Imaging Enables Mapping of Matrix Architectures that Promote Pancreatic Ductal Adenocarcinoma Dissemination. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00160-9. [PMID: 40350060 DOI: 10.1016/j.ajpath.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 02/22/2025] [Accepted: 04/08/2025] [Indexed: 05/14/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is an extremely metastatic and lethal disease. In PDA, extracellular matrix (ECM) architectures, known as tumor-associated collagen signatures (TACSs), regulate invasion and metastatic spread in both early dissemination and late-stage disease. As such, TACS has been suggested as a biomarker to aid in pathologic assessment. However, despite its significance, approaches to quantitatively capture these ECM patterns currently require advanced optical systems with signaling processing analysis. Here, we present an expansion of polychromatic polarized microscopy (PPM) with inherent angular information coupled to machine learning and computational pixel-wise analysis of TACS. Using this platform, we are able to accurately capture TACS architectures in hematoxylin and eosin-stained histology sections directly through PPM contrast. Moreover, PPM facilitated identification of transitions to dissemination architectures (ie, transitions from sequestration through expansion to dissemination from both PanINs and throughout PDA). Last, PPM evaluation of architectures in liver metastases, the most common metastatic site for PDA, demonstrates TACS-mediated focal and local invasion as well as identification of unique patterns anchoring aligned fibers into normal-adjacent tumor, suggesting that these patterns may be precursors to metastasis expansion and local spread from micrometastatic lesions. Combined, these findings demonstrate that PPM coupled to computational platforms is a powerful tool for analyzing ECM architecture that can be used to advance cancer microenvironment studies and provide clinically relevant diagnostic information.
Collapse
Affiliation(s)
- Guhan Qian
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; Center for Multiparametric Imaging of Tumor Immune Microenvironments, University of Minnesota and University of Wisconsin-Madison
| | - Hongrong Zhang
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; Center for Multiparametric Imaging of Tumor Immune Microenvironments, University of Minnesota and University of Wisconsin-Madison
| | - Yuming Liu
- Center for Multiparametric Imaging of Tumor Immune Microenvironments, University of Minnesota and University of Wisconsin-Madison; Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, Wisconsin
| | - Michael Shribak
- Marine Biological Laboratory, University of Chicago, Woods Hole, Massachusetts
| | - Kevin W Eliceiri
- Center for Multiparametric Imaging of Tumor Immune Microenvironments, University of Minnesota and University of Wisconsin-Madison; Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, Wisconsin; Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Paolo P Provenzano
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; Center for Multiparametric Imaging of Tumor Immune Microenvironments, University of Minnesota and University of Wisconsin-Madison; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota; Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Institute for Engineering in Medicine, University of Minnesota, Minneapolis, Minnesota; Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
25
|
Preckwinkel P, Mir KUI, Otto FW, Elrewany H, Sinz A, Hüttelmaier S, Bley N, Gutschner T. Long Non-Coding RNAs and RNA-Binding Proteins in Pancreatic Cancer Development and Progression. Cancers (Basel) 2025; 17:1601. [PMID: 40427100 PMCID: PMC12110025 DOI: 10.3390/cancers17101601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer and is responsible for about 467,000 cancer deaths annually. An oftentimes asymptomatic early phase of this disease results in a delayed diagnosis, and patients often present with advanced disease. Current treatment options have limited survival benefits, and only a minor patient population carries actionable genomic alterations. Hence, innovative personalized treatment strategies that consider molecular, cellular and functional analyses are urgently needed for pancreatic cancer patients. However, the majority of the genetic alterations found in PDAC are currently undruggable, or patients' response is not as expected. Therefore, non-genomic biomarkers and alternative molecular targets should be considered in order to advance the clinical management of PDAC patients. In line with this, recent gene expression and single-cell transcriptome analyses have identified molecular subtypes and transcriptional cell states that affect disease progression and drug efficiency. In this review, we will introduce long non-coding RNAs (lncRNAs) as well as RNA-binding proteins (RBPs) that are able to modulate the transcriptome of a cell through diverse mechanisms, thereby contributing to disease progression. We will provide a brief overview about the general functions of lncRNAs and RBPs, respectively. Subsequently, we will highlight selected lncRNAs and RBPs that have been shown to play a role in PDAC development, progression and drug response. Finally, we will present strategies aiming to interfere with the expression and function of lncRNAs and RBPs.
Collapse
Affiliation(s)
- Pit Preckwinkel
- Section for RNA Biology and Pathogenesis, Institute of Molecular Medicine, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Khursheed Ul Islam Mir
- Section for Molecular Cell Biology, Institute of Molecular Medicine, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (K.U.I.M.); (H.E.); (S.H.)
| | - Florian W. Otto
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (F.W.O.); (A.S.)
- Center for Structural Mass Spectrometry, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Hend Elrewany
- Section for Molecular Cell Biology, Institute of Molecular Medicine, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (K.U.I.M.); (H.E.); (S.H.)
| | - Andrea Sinz
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (F.W.O.); (A.S.)
- Center for Structural Mass Spectrometry, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Stefan Hüttelmaier
- Section for Molecular Cell Biology, Institute of Molecular Medicine, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (K.U.I.M.); (H.E.); (S.H.)
| | - Nadine Bley
- Section for Molecular Cell Biology, Institute of Molecular Medicine, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (K.U.I.M.); (H.E.); (S.H.)
| | - Tony Gutschner
- Section for RNA Biology and Pathogenesis, Institute of Molecular Medicine, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| |
Collapse
|
26
|
Glapiński F, Zając W, Fudalej M, Deptała A, Czerw A, Sygit K, Kozłowski R, Badowska-Kozakiewicz A. The Role of the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma: Recent Advancements and Emerging Therapeutic Strategies. Cancers (Basel) 2025; 17:1599. [PMID: 40427098 PMCID: PMC12110676 DOI: 10.3390/cancers17101599] [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: 04/07/2025] [Revised: 04/30/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic cancer (PC), with pancreatic ductal adenocarcinoma (PDAC) comprising about 90% of all cases, is one of the most aggressive and lethal solid tumors. PDAC remains one of the most significant challenges of oncology to this day due to its inadequate response to conventional treatment, gradual rise in incidence since 2004, and poor five-year survival rates. As cancer cells are the primary adversary in this uneven fight, they remain the primary research target. Nevertheless, increasing attention is being paid to the tumor microenvironment (TME). The most crucial TME constellation components are immune cells, especially macrophages, stellate cells and lymphocytes, fibroblasts, bacterial and fungal microflora, and neuronal cells. Depending on the particular phenotype of these cells, the composition of the microenvironment, and the cell ratio, patients can experience different disease outcomes and varying vulnerability to treatment approaches. This study aims to present the current knowledge and review the most up-to-date scientific findings regarding the microenvironment of PC. It contains detailed information on the structure and cellular composition of the stroma, including its impact on disease development, metastasis, and response to treatment, as well as the therapeutic opportunities that arise from targeting this tissue.
Collapse
Affiliation(s)
- Franciszek Glapiński
- Students’ Scientific Organization of Cancer Cell Biology, Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Weronika Zając
- Students’ Scientific Organization of Cancer Cell Biology, Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Marta Fudalej
- Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.F.); (A.D.)
- Department of Oncology, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
| | - Andrzej Deptała
- Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.F.); (A.D.)
| | - Aleksandra Czerw
- Department of Health Economics and Medical Law, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Department of Economic and System Analyses, National Institute of Public Health NIH—National Research Institute, 00-791 Warsaw, Poland
| | - Katarzyna Sygit
- Faculty of Health Sciences, Calisia University, 62-800 Kalisz, Poland
| | - Remigiusz Kozłowski
- Department of Management and Logistics in Healthcare, Medical University of Lodz, 90-131 Lodz, Poland
| | - Anna Badowska-Kozakiewicz
- Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.F.); (A.D.)
| |
Collapse
|
27
|
Bolm L, Fraunhoffer N, Dusetti N, Straesser J, Petruch N, Fernandez del Castillo C, Iovanna J. The PancreasView gemcitabine transcriptomic signature predicts response to gemcitabine in patients with resected pancreatic ductal adenocarcinoma. Oncologist 2025; 30:oyaf083. [PMID: 40349133 PMCID: PMC12065937 DOI: 10.1093/oncolo/oyaf083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/28/2025] [Indexed: 05/14/2025] Open
Abstract
INTRODUCTION In the current study, we aimed to assess the efficacy of a gemcitabine response predictive signature that is part of the PancreasView transcriptomic predictive tool (Gem + or Gem-). METHODS We used a cohort of pancreatic ductal adenocarcinoma patients treated from the Massachusetts General Hospital who underwent upfront resection. RESULTS In this cohort of 43 patients, 20 (46.5%) received adjuvant gemcitabine (GEM arm) and 23 (53.5%) did not receive any adjuvant chemotherapy. Among the 43 patients, the Gem signature defined a subgroup of 16 patients (37.2%) who were sensitive (Gem+) and 27 (62.8%), who were resistant to gemcitabine (Gem-). The Gem+ patients who received adjuvant gemcitabine had significantly better median disease-free survival (DFS) compared to the Gem- patients (NR until 72 months of follow-up vs 19.0 months; stratified hazard ratio [HR]: 0.19; 95% CI, 0.04-0.86; P = .032) and longer median cancer-specific survival (CSS) (NR until 96 months of follow-up vs 37.0 months; stratified HR: 0.18; 95% CI, 0.04-0.85; P = .030) when treated with gemcitabine. The gemcitabine signature remained an independent predictive factor for DFS (HR: 0.41; 95% CI, 0.19-0.89; P = .024) and CSS (HR: 0.47; 95% CI, 0.22-1.23; P = .059) after adjusting for clinicopathological characteristics in an unstratified univariate Cox hazard model. CONCLUSIONS This validation of the gemcitabine predictive transcriptomic signature in an independent cohort from Massachusetts General Hospital reinforces the robustness and reliability of this tool. This study highlights the potential of the signature to aid in the personalization of chemotherapy and enhance patient outcomes in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Louisa Bolm
- Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, United States
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck 23562, Germany
| | - Nicolas Fraunhoffer
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix- Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille 13288, France
- Center for Pharmacological and Botanical Studies, Faculty of Medicine, Buenos Aires University, National Council for Scientific and Technical Research, Buenos Aires C1121ABG, Argentina
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix- Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille 13288, France
| | - Julia Straesser
- Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, United States
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck 23562, Germany
| | - Natalie Petruch
- Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, United States
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck 23562, Germany
| | | | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix- Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille 13288, France
- Gastroenterology Section, Hospital de Alta Complejidad El Cruce, Florencio Varela, Buenos Aires, 1888, Argentina
- School of Medicine, University Arturo Jauretche, Florencio Varela, Buenos Aires, 1888, Argentina
| |
Collapse
|
28
|
Belfiori G, De Stefano F, Tamburrino D, Gasparini G, Aleotti F, Camisa PR, Arcangeli C, Schiavo Lena M, Pecorelli N, Palumbo D, Partelli S, De Cobelli F, Reni M, Crippa S, Falconi M. Anatomically resectable versus biologically borderline resectable pancreatic cancer definition: refining the border beyond anatomical criteria and biological aggressiveness. BJS Open 2025; 9:zraf033. [PMID: 40392528 PMCID: PMC12090895 DOI: 10.1093/bjsopen/zraf033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND The anatomically resectable pancreatic ductal adenocarcinoma treatment sequence is still debated. Heterogeneity in patient characteristics within this group may explain literature discrepancies. To overcome these limits, a biologically borderline resectable pancreatic ductal adenocarcinoma category has been analysed according to institutional criteria. The aim of this study was to examine the characteristics and outcomes of patients with biologically borderline resectable pancreatic ductal adenocarcinoma and determine whether they represent a distinct clinical and prognostic subgroup. METHODS Data from all consecutive patients who underwent surgical resection for pancreatic ductal adenocarcinoma between 2015 and 2022 were retrospectively analysed. Biologically borderline resectable disease was classified by the presence of one or more of the following: carbohydrate antigen 19-9 ≥200 U/ml, cancer-related symptoms lasting >40 days, and radiological suspicion of regional lymph node metastases at diagnosis. RESULTS In total, 886 patients were included in the study and divided into anatomically borderline resectable (266 patients (30%)) and anatomically resectable (620 patients (70%)), which was further divided into resectable (R; 397 patients (64%)) and biologically borderline resectable (223 patients (36%)). Neoadjuvant treatment was administered in 245 patients (92.1%) in the anatomically borderline resectable group, 82 patients (20.7%) in the R group, and 135 patients (60.5%) in the biologically borderline resectable group. After a median follow-up of 45 (95% c.i. 42 to 48) months, the median disease-specific survival in the biologically borderline resectable group was 40 months compared with 59 months in the R group (P < 0.001) and 40 months in the anatomically borderline resectable group (P = 0.570). In the upfront surgery cohort, the median disease-specific survival was worse for biologically borderline resectable patients compared with R patients (27 versus 54 months respectively, P < 0.001). Biologically borderline resectable was also independently associated with worse disease-specific survival, together with age, tumour size at diagnosis, and anatomically borderline resectable. The same, except for age, were also predictors of worse event-free survival. CONCLUSION Despite their identical anatomical appearance, resectable and biologically borderline resectable pancreatic ductal adenocarcinoma represent two distinct prognostic entities, warranting separate evaluation and, potentially, different treatment approaches.
Collapse
Affiliation(s)
- Giulio Belfiori
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federico De Stefano
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Domenico Tamburrino
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Gasparini
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Francesca Aleotti
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo R Camisa
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Claudia Arcangeli
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Marco Schiavo Lena
- Department of Pathology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicolo Pecorelli
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Diego Palumbo
- Department of Radiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Partelli
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco De Cobelli
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Radiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Michele Reni
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Crippa
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Falconi
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
29
|
Stoop TF, Wu YHA, Oba A, Ali M, Feld IM, Al-Musawi MH, Jain A, Saiura A, Sauvanet A, Coppola A, Javed AA, Groot Koerkamp B, Miller BN, Hashimoto D, Caputo D, Kleive D, Sereni E, Kazemier G, Belfiori G, Ishida H, van Dam JL, Dembinski J, Akahoshi K, Roberts KJ, Tanaka K, Labori KJ, Falconi M, House MG, Sugimoto M, Tanabe M, Gotohda N, Chatzizacharias N, Krohn PS, Dokmak S, Rodriguez Franco S, Hirano S, Burgdorf SK, Crippa S, Satoi S, Nguyen TK, Yamamoto T, Nakamura T, Ushida Y, Bachu V, Burns WR, Inoue Y, Takahashi Y, Aslami Z, Schulick RD, He J, Messersmith W, Besselink MG, Burkhart RA, Wilmink JW, Del Chiaro M. Survival after neoadjuvant and induction FOLFIRINOX versus gemcitabine-nab-paclitaxel in patients with resected localised pancreatic adenocarcinoma: an international multicentre study. Br J Cancer 2025:10.1038/s41416-025-03025-1. [PMID: 40328917 DOI: 10.1038/s41416-025-03025-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 03/15/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND It remains unclear whether there is a difference in overall survival (OS) benefit between (m)FOLFIRINOX and gemcitabine-nab-paclitaxel as preoperative regimens for localised pancreatic adenocarcinoma. This study aimed to investigate the outcome of patients with resected localised pancreatic adenocarcinoma following (m)FOLFIRINOX versus gemcitabine-nab-paclitaxel. METHODS International multicentre retrospective study (16 centres, 8 countries, 3 continents), including consecutive patients after pancreatic resection for localised pancreatic adenocarcinoma following 2-6 months preoperative (m)FOLFIRINOX or gemcitabine-nab-paclitaxel (2010-2018). Primary endpoint was OS from start of preoperative chemotherapy. Cox regression analysis was performed to investigate the association of the preoperative chemotherapy regimen with OS, adjusted for confounders at diagnosis. RESULTS Overall, 935 patients were included after resection of localised pancreatic adenocarcinoma following preoperative (m)FOLFIRINOX (65%) or gemcitabine-nab-paclitaxel (35%). Preoperative chemotherapy regimen (m)FOLFIRINOX was not associated with OS (HR = 0.83 [95% CI 0.64-1.08]), compared to gemcitabine-nab-paclitaxel. Interaction analysis showed stronger effect of (m)FOLFIRINOX in patients with a lower (i.e., non-elevated/marginally elevated) serum CA19-9 at diagnosis (pinteraction = 0.032). CONCLUSION This international study found no OS benefit of preoperative (m)FOLFIRINOX in patients with resected localised pancreatic adenocarcinoma compared to gemcitabine-nab-paclitaxel.
Collapse
Affiliation(s)
- Thomas F Stoop
- Division of Surgical Oncology, Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Amsterdam, The Netherlands.
| | - Y H Andrew Wu
- Division of Surgical Oncology, Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Atsushi Oba
- Division of Surgical Oncology, Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | - Mahsoem Ali
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, The Netherlands
| | - Isabel M Feld
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Mohammed H Al-Musawi
- Clinical Trials of Office, Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Ajay Jain
- Division of Surgical Oncology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Akio Saiura
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Alain Sauvanet
- Department of HPB Surgery and Liver Transplantation, Hôpital Beaujon, University Paris Cité, INSERM Unité Mixte de Recherche 1149, Clichy, France
| | | | - Ammar A Javed
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Division of Surgical Oncology, Department of Surgery, New York University Medical Center, New York City, NY, USA
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Braden N Miller
- Division of Surgical Oncology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Damiano Caputo
- Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of General Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Dyre Kleive
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Elisabetta Sereni
- Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Department of General and Pancreatic Surgery, The Pancreas Institute University of Verona, Hospital Trust, Verona, Italy
| | - Geert Kazemier
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, The Netherlands
| | - Giulio Belfiori
- Pancreatic Surgery, IRCCS San Raffaele Hospital, Vita-Salute University, Milano, Italy
| | - Hirofumi Ishida
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Jacob L van Dam
- Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jeanne Dembinski
- Department of HPB Surgery and Liver Transplantation, Hôpital Beaujon, University Paris Cité, INSERM Unité Mixte de Recherche 1149, Clichy, France
| | - Keiichi Akahoshi
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keith J Roberts
- Hepato-Pancreato-Biliary Unit, Department of Surgery, University Hospitals of Birmingham, Birmingham, UK
| | - Kimitaka Tanaka
- Department of Gastroenterological Surgery II, Hokkaido University, Faculty of Medicine, Hokkaido, Japan
| | - Knut J Labori
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Massimo Falconi
- Pancreatic Surgery, IRCCS San Raffaele Hospital, Vita-Salute University, Milano, Italy
| | - Michael G House
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Motokazu Sugimoto
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Minoru Tanabe
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Naoto Gotohda
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Nikolaos Chatzizacharias
- Hepato-Pancreato-Biliary Unit, Department of Surgery, University Hospitals of Birmingham, Birmingham, UK
| | - Paul S Krohn
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Safi Dokmak
- Department of HPB Surgery and Liver Transplantation, Hôpital Beaujon, University Paris Cité, INSERM Unité Mixte de Recherche 1149, Clichy, France
| | - Salvador Rodriguez Franco
- Division of Surgical Oncology, Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Hokkaido University, Faculty of Medicine, Hokkaido, Japan
| | - Stefan K Burgdorf
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Stefano Crippa
- Pancreatic Surgery, IRCCS San Raffaele Hospital, Vita-Salute University, Milano, Italy
| | - Sohei Satoi
- Division of Surgical Oncology, Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Department of Surgery, Kansai Medical University, Osaka, Japan
| | - Trang K Nguyen
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tomohisa Yamamoto
- Department of Gastroenterological Surgery II, Hokkaido University, Faculty of Medicine, Hokkaido, Japan
| | - Toru Nakamura
- Department of Gastroenterological Surgery II, Hokkaido University, Faculty of Medicine, Hokkaido, Japan
| | - Yuta Ushida
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | - Vismaya Bachu
- Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - William R Burns
- Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Yosuke Inoue
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | - Yu Takahashi
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | - Zohra Aslami
- Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Richard D Schulick
- Division of Surgical Oncology, Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Jin He
- Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Wells Messersmith
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marc G Besselink
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Richard A Burkhart
- Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Johanna W Wilmink
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Oncology, Amsterdam, The Netherlands
| | - Marco Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
30
|
Akkus E, Lamarca A. Adjuvant chemotherapy compared to observation in resected biliary tract cancers: Survival meta-analysis of phase-III randomized controlled trials. Eur J Cancer 2025; 220:115342. [PMID: 40101432 DOI: 10.1016/j.ejca.2025.115342] [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: 01/22/2025] [Revised: 02/23/2025] [Accepted: 03/03/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND A limited number of randomized controlled trials (RCTs) investigated adjuvant chemotherapy in biliary tract cancers (BTCs). Recurrences and deaths are common in the first 2 years and survival remains poor despite adjuvant treatment. METHODS Phase-III RCTs were included comparing adjuvant chemotherapy and observation in resected BTCs. The primary endpoints were recurrence-free (RFS) and overall survival (OS). Proportional hazard results were used for trial-based analyses. Patient data was curated from published Kaplan-Meier curves to analyze short-term (2-year) hazards. The Parmar and generic inverse variance methods were used. RESULTS 1308 patients in 4 trials (BILCAP, ASCOT, BCAT, PRODIGE-12) were included. Capecitabine (BILCAP) and S-1 (ASCOT) were grouped as 5-FU-based, gemcitabine (BCAT) and gemcitabine-oxaliplatin (PRODIGE-12) were grouped as gemcitabine-based chemotherapy. Adjuvant 5FU-based chemotherapy improved RFS [HR: 0.80 (95 % CI:0.68-0.95), p = 0.012] and OS [HR: 0.78 (95 % CI:0.65-0.94), p = 0.009]. However, gemcitabine-based chemotherapy did not provide benefit in RFS [HR: 0.90 (95 % CI:0.70-1.15), p = 0.428] and OS [HR: 1.03 (95 % CI:0.78-1.36), p = 0.794]. The benefit of 5-FU-based chemotherapy was more apparent in the short-term (2-year hazards) (RFS: [HR: 0.67 (95 %CI:57-0.79), p < 0.001] and OS: [HR: 0.61 (95 % CI:59-0.64), p < 0.001]). However, gemcitabine-based chemotherapy did not provide RFS benefit in the short term either [HR: 0.80 (95 % CI:0.64-0.1.01), p = 0.067] and seemed to be even detrimental for OS [HR: 1.22 (95 % CI:1.14-1.31), p < 0.001] in the first 2 years. CONCLUSION This study confirms using 5FU-based monotherapy in the adjuvant treatment of resected BTCs. The more prominent benefit in the first 2 years emphasizes that more effective adjuvant treatments with sustained long-term benefits are needed. Two-year proportional hazards OS and RFS are proposed here as an additional secondary end-point to consider in future clinical trials. in this setting. Registration ID (PROSPERO): CRD42024614444.
Collapse
Affiliation(s)
- Erman Akkus
- Ankara University Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye; Ankara University Cancer Research Institute, Ankara, Türkiye
| | - Angela Lamarca
- Department of Oncology, OncoHealth Institute, Instituto de Investigaciones Sanitarias FJD, Fundación Jiménez Díaz University Hospital, Madrid, Spain.
| |
Collapse
|
31
|
Graus MUJE, van Diepen AE, Josemanders K, Besselink MG, Bouwense SAW, Daamen LA, de Hingh IHJT, de Jong EJM, van Laarhoven HWM, de Meijer VE, Quintus Molenaar I, Stommel MWJ, Valkenburg-van Iersel LBJ, Wilmink JW, van der Geest LGM, de Vos-Geelen J. Patterns of disease dissemination and survival in patients with synchronous and metachronous metastatic pancreatic adenocarcinoma: Nationwide population-based study. Eur J Cancer 2025; 220:115385. [PMID: 40154212 DOI: 10.1016/j.ejca.2025.115385] [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: 01/16/2025] [Revised: 03/11/2025] [Accepted: 03/16/2025] [Indexed: 04/01/2025]
Abstract
AIM Despite advances in understanding pancreatic adenocarcinoma, evidence on its metastatic patterns and impact on patient survival remains limited. This study aims to identify patterns of disease dissemination in synchronous versus metachronous metastatic pancreatic adenocarcinoma (mPAC) and their association with overall survival (OS). METHODS Patients diagnosed with synchronous- or metachronous-mPAC were selected from the Netherlands Cancer Registry (2015-2021). Patient, tumor, and treatment characteristics were compared using Chi-squared tests. Survival data, calculated from detection of metastatic disease (OS-M), were analyzed using Kaplan-Meier and Log-rank tests. RESULTS Overall, 10,788 patients with synchronous- and 508 with metachronous-mPAC were included. Median time to first metastasis in metachronous-mPAC was 13.2 months (IQR 9-23), varying significantly by metastatic site (liver-only 11.5; lung-only 28.0 months). Compared to synchronous-mPAC, patients with metachronous-mPAC had less liver metastases (48 % versus 75 %, p < 0.001), but more lung (29 % versus 21 %, p < 0.001) and peritoneal (35 % versus 25 %, p < 0.001) metastases. Synchronous metastases to liver-only, lung-only, lymph node-only, or multiple sites at first diagnosis had a median OS-M that was (nearly) half compared to metachronous metastases to the same sites. Bone-only or peritoneum-only metastases in synchronous-mPAC showed a median OS-M comparable to metachronous-mPAC. CONCLUSION This nationwide population-based study reveals that metachronous-mPAC less commonly presents with liver metastases and more often metastasizes to lung, peritoneum or other atypical sites compared to synchronous-mPAC. These distinct metastatic patterns and their differences in survival may help enhance the prognostic estimation for individual patients from the detection of metastatic disease and warrants further research into the biology underlying metastasis development.
Collapse
Affiliation(s)
- Merlijn U J E Graus
- Maastricht University Medical Center, Department of Internal Medicine, Division of Medical Oncology, Maastricht, the Netherlands; GROW - Research institute for Oncology & Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Aniek E van Diepen
- Maastricht University Medical Center, Department of Internal Medicine, Division of Medical Oncology, Maastricht, the Netherlands; GROW - Research institute for Oncology & Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Kim Josemanders
- Canisius Wilhelmina Hospital (CWZ), Department oF Internal Medicine, Nijmegen, the Netherlands
| | - Marc G Besselink
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| | - Stefan A W Bouwense
- Maastricht University Medical Center, Department of Surgery, NUTRIM - Institute of Nutrition and Translational Research in Metabolism, Maastricht, the Netherlands
| | - Lois A Daamen
- University Medical Center Utrecht, Division of Imaging and Oncology, Utrecht, the Netherlands
| | - Ignace H J T de Hingh
- GROW - Research institute for Oncology & Reproduction, Maastricht University, Maastricht, the Netherlands; Catharina Hospital Eindhoven, Department of Surgery, Eindhoven, the Netherlands
| | - Evelien J M de Jong
- Maastricht University Medical Center, Department of Internal Medicine, Division of Medical Oncology, Maastricht, the Netherlands; GROW - Research institute for Oncology & Reproduction, Maastricht University, Maastricht, the Netherlands; Zuyderland Medical Center, Department of Internal Medicine, Sittard-Geleen, the Netherlands
| | - Hanneke W M van Laarhoven
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands; Amsterdam UMC, Location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands
| | - Vincent E de Meijer
- University Medical Center Groningen, Department of Surgery, Section of Hepatobiliary Surgery and Liver Transplantation, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, the Netherlands
| | - I Quintus Molenaar
- University Medical Center Utrecht, Division of Imaging and Oncology, Utrecht, the Netherlands
| | - Martijn W J Stommel
- Radboud University Medical Center, Department of Surgery, Nijmegen, the Netherlands
| | - Liselot B J Valkenburg-van Iersel
- Maastricht University Medical Center, Department of Internal Medicine, Division of Medical Oncology, Maastricht, the Netherlands; GROW - Research institute for Oncology & Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Johanna W Wilmink
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands; Amsterdam UMC, Location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands
| | - Lydia G M van der Geest
- Netherlands Comprehensive Cancer Organisation (IKNL), Department of Research, Utrecht, the Netherlands
| | - Judith de Vos-Geelen
- Maastricht University Medical Center, Department of Internal Medicine, Division of Medical Oncology, Maastricht, the Netherlands; GROW - Research institute for Oncology & Reproduction, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
32
|
Patel S, Jenkins E, Kusurkar RP, Lee S, Jiang W, Nevler A, McCoy M, Pishvaian MJ, Sears RC, Brody JR, Yeo CJ, Jain A. Targeting BARD1 suppresses a Myc-dependent transcriptional program and tumor growth in pancreatic ductal adenocarcinoma. Neoplasia 2025; 63:101152. [PMID: 40096771 PMCID: PMC11957605 DOI: 10.1016/j.neo.2025.101152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest cancers demanding better and more effective therapies. BARD1 or BRCA1-Associated -Ring Domain-1 plays a pivotal role in homologous recombination repair (HRR). However, its function and the underlying molecular mechanisms in PDAC are still not fully elucidated. Here, we demonstrate that BARD1 is overexpressed in PDAC and its genetic inhibition suppresses c-Myc and disrupts c-Myc dependent transcriptional program. Mechanistically, BARD1 stabilizes c-Myc through ubiquitin-proteasome system by regulating FBXW7. Importantly, targeting BARD1 using either siRNAs or CRISPR/Cas9 deletion blocks PDAC growth in vitro and in vivo, without any signs of toxicity to mice. Using a focused drug library of 477 DNA damage response compounds, we also found that BARD1 inhibition enhances therapeutic efficacy of several clinically relevant agents (fold changes ≥4), including PARPi, in HRR proficient PDAC cells. These data uncover BARD1 as an attractive therapeutic target for HRR proficient PDAC.
Collapse
Affiliation(s)
- Sohum Patel
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Eleanor Jenkins
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rutuj P Kusurkar
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sherry Lee
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Wei Jiang
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Avinoam Nevler
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Matthew McCoy
- Innovation Center for Biomedical Informatics & Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Michael J Pishvaian
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rosalie C Sears
- Department of Molecular and Medical Genetics, and Brenden-Colson Center for Pancreatic Care Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Jonathan R Brody
- Department of Surgery, and Brenden-Colson Center for Pancreatic Care Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Charles J Yeo
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Aditi Jain
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
33
|
Kuijper SC, Gehrels AM, van der Geest LG, Verhoeven RHA, Koerkamp BG, Molenaar IQ, Stommel MWJ, de Meijer VE, de Vos‐Geelen J, Wumkes ML, Besselink MG, Wilmink JW, van Laarhoven HWM, for the Dutch Pancreatic Cancer Group. Survival scenarios of patients with localized and metastatic pancreatic adenocarcinoma: A population-based study. Int J Cancer 2025; 156:1726-1735. [PMID: 39614657 PMCID: PMC11887001 DOI: 10.1002/ijc.35267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 12/01/2024]
Abstract
Pancreatic adenocarcinoma (PAC) is notorious for its poor survival. The provision of survival scenarios-that is, best-case, typical and worst-case scenarios-could prove valuable to patients and clinicians. This study investigated survival scenarios and how these have changed over a period of 16 years for patients with PAC. Data from the Netherlands Cancer Registry were used to identify patients with localized and metastatic PAC (2005-2021). Survival scenarios, including best-case, upper-typical, typical (median), lower-typical, and worst-case, were estimated based on survival curve percentiles (p10, p25, p50, p75, and p90). Annual differences were assessed for significance using weighted linear regression analyses. Factors associated with these scenarios were identified through univariable tests. Overall, 14,622 patients with localized and 20,199 with metastatic PAC were included. For patients with localized PAC, the best, upper-typical and typical survival scenarios improved statistically significant with average annual improvement of 1.54 (95%CI: 1.2-1.88), 0.67 (0.56-0.78), and 0.24 (0.19-0.29) months, respectively. For patients with metastatic PAC the best and upper-typical survival scenarios increased statically significantly with annual improvement of 0.28 (0.21-0.34) and 0.06 (0.02-0.09) months, respectively. The best-case and upper-typical scenarios were associated with younger patients, more aggressive disease-focused treatments, fewer comorbidities, and better overall performance status. Over the past 16 years, survival improvements in patients with PAC have been most notable in these scenarios. Although the absolute gains were modest, these results offer encouraging potential for advancements in life-prolonging care for this type of cancer.
Collapse
Affiliation(s)
- Steven C. Kuijper
- Department of Medical OncologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer Treatment and Quality of LifeAmsterdamThe Netherlands
| | - Anne M. Gehrels
- Department of Medical OncologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer Treatment and Quality of LifeAmsterdamThe Netherlands
| | - Lydia G. van der Geest
- Department of Medical OncologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer Treatment and Quality of LifeAmsterdamThe Netherlands
- Department of Research & DevelopmentNetherlands Comprehensive Cancer Organisation (IKNL)UtrechtThe Netherlands
| | - Rob H. A. Verhoeven
- Department of Medical OncologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer Treatment and Quality of LifeAmsterdamThe Netherlands
- Department of Research & DevelopmentNetherlands Comprehensive Cancer Organisation (IKNL)UtrechtThe Netherlands
| | - Bas Groot Koerkamp
- Department of SurgeryErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - I. Quintus Molenaar
- Department of Surgery, Regional Academic Cancer Center UtrechtUniversity Medical Center Utrecht and St. Antonius Hospital NieuwegeinUtrechtThe Netherlands
| | | | - Vincent E. de Meijer
- Department of SurgeryUniversity of Groningen and University Medical Center GroningenGroningenThe Netherlands
| | - J. de Vos‐Geelen
- Department of Internal Medicine, Division of Medical OncologyMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Miriam L. Wumkes
- Department of Medical OncologyJeroen Bosch HospitalDen BoschThe Netherlands
| | - Marc G. Besselink
- Cancer Center AmsterdamCancer Treatment and Quality of LifeAmsterdamThe Netherlands
- Amsterdam UMC, location University of AmsterdamDepartment of SurgeryAmsterdamThe Netherlands
| | - Johanna W. Wilmink
- Department of Medical OncologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer Treatment and Quality of LifeAmsterdamThe Netherlands
| | - Hanneke W. M. van Laarhoven
- Department of Medical OncologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer Treatment and Quality of LifeAmsterdamThe Netherlands
| | | |
Collapse
|
34
|
He Y, Zhu Y, Wang W, Yi Y, Wang Z, Zhao C, Li J, Huang X, Zheng L. Clinical efficacy and chemoresistance analysis of precision neoadjuvant chemotherapy for borderline resectable pancreatic cancer: a prospective, single-arm pilot study. Int J Surg 2025; 111:3269-3280. [PMID: 40146255 PMCID: PMC12165568 DOI: 10.1097/js9.0000000000002342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/06/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Neoadjuvant chemotherapy (NAC) can improve the survival outcomes of patients with pancreatic cancer, but for borderline resectable pancreatic cancer (BRPC) the proportion of conversion to surgery remains unsatisfactory. This single-arm pilot study aimed to assess the clinical efficacy and safety of NAC based on patient-derived organoids (PDOs) for BRPC. METHODS Biopsy samples from BRPC patients were collected for generating PDOs. Gemcitabine plus nab-paclitaxel as NAC was initially administrated for one cycle, and then the treatment regimen was adjusted based on the PDO drug sensitivity testing. The primary endpoint was the objective response rate (ORR). Secondary endpoints included R0 resection rate, NAC-related adverse events (AEs), and postoperative complications. Exploratory objectives were to assess the chemoresistance to gemcitabine. RESULTS Totally 19 of 25 patients were eligible for the study, among whom 16 achieved partial response and received surgical resection, with the ORR of 84.2% (16/19). The R0 resection rate was 81.3% (13/16). During NAC, 8 (42.1%, 8/19) patients experienced different grades of AEs, mainly including grade 2 myelosuppression (26.3%), cutaneous pruritus (5.3%), and diarrhea (5.3%). scRNA-seq analysis of duct cells showed that the transcriptome in aneuploid cells may affect gemcitabine resistance via multiple pathways, among which upregulation of drug-resistant genes ( OLFM4, AGR2, MUC5AC, MUC1, HMGA1, REG4, IL17RB, GCNT3, AKR1B10, ITGA6, HMGCS2 , and SQLE ) and downregulation of sensitive genes ( SIK1, HEXIM1, SPINT2, GADD45 , and TIMP2 ) played crucial roles. Changes in the interactions between cancer cells and other cell groups may also involve in gemcitabine resistance. CONCLUSION PDO-based NAC shows a promising resectable rate in BRPC patients, with good tolerance. Potential drug-resistant and sensitive genes and cell-cell interaction changes may participate in the development of gemcitabine resistance.
Collapse
Affiliation(s)
- Yonggang He
- Department of Hepatobiliary, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yinan Zhu
- Department of Hepatobiliary, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Weiwei Wang
- Department of Hepatobiliary and Pancreatic Surgery, Chongqing Tongliang District People’s Hospital, Chongqing, China
| | - Yuanyue Yi
- Department of Pathology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Zheng Wang
- Department of Hepatobiliary, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Chongyu Zhao
- Department of Hepatobiliary, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Jing Li
- Department of Hepatobiliary, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xiaobing Huang
- Department of Hepatobiliary, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Lu Zheng
- Department of Hepatobiliary, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
35
|
D'Ambra V, Ricci C, Ingaldi C, Alberici L, Minghetti M, Casadei R. Stage IV pancreatic ductal adenocarcinoma (PDAC) with synchronous liver metastasis: are there survival benefits in liver resection? A systematic review and meta-analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109598. [PMID: 39837137 DOI: 10.1016/j.ejso.2025.109598] [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: 09/25/2024] [Revised: 01/01/2025] [Accepted: 01/10/2025] [Indexed: 01/23/2025]
Abstract
OBJECTIVE Metastatic PDAC has a very poor prognosis, and surgery has a limited role. The study aims to evaluate the OS of patients with PDAC and synchronous liver metastasis who undergo surgical therapy (ST) versus non-surgical therapies (NST). METHODS We performed a random effects meta-analysis. Inclusion criteria were: PDAC histology; studies reporting technically resectable cases with liver metastasis and survival data; absence of extra-hepatic disease. The primary endpoint was to evaluate OS. Results were reported as HR and 95 % CI. We performed a meta-regression analysis to identify factors influencing heterogeneity. We analyzed key covariates in order to predict how changes in these factors affect HR. RESULTS Six studies were included. The OS was significantly better in group ST than NST, with HR = 0.41 (95 % CI: 0.32-0.52). Heterogeneity was high (I2 = 64.50 %). As the rate of patients who underwent postoperative CT in the ST group decreased, the difference between the two groups decreased (β = -1.28 ± 0.67; p = 0.003), with almost 87.10 % heterogeneity. The adjusted effect based on meta-regression showed an improved OS in ST group only when both pre- and post-operative systemic CT were administrated (HR 0.18, 95 % CI: 0.08-0.40). CONCLUSIONS In highly selected patients with metastatic PDAC who respond to systemic CT and receive post-operative systemic CT, ST could be associated with improved OS. However, the high heterogeneity and retrospective design of included studies limit the ability to draw definitive conclusions.
Collapse
Affiliation(s)
- Vincenzo D'Ambra
- Division of Pancreatic Surgery, IRCCS, Azienda Ospedaliero Universitaria di Bologna, Italy; Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, S. Orsola-Malpighi Hospital, Italy
| | - Claudio Ricci
- Division of Pancreatic Surgery, IRCCS, Azienda Ospedaliero Universitaria di Bologna, Italy; Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, S. Orsola-Malpighi Hospital, Italy.
| | - Carlo Ingaldi
- Division of Pancreatic Surgery, IRCCS, Azienda Ospedaliero Universitaria di Bologna, Italy; Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, S. Orsola-Malpighi Hospital, Italy
| | - Laura Alberici
- Division of Pancreatic Surgery, IRCCS, Azienda Ospedaliero Universitaria di Bologna, Italy; Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, S. Orsola-Malpighi Hospital, Italy
| | - Margherita Minghetti
- Division of Pancreatic Surgery, IRCCS, Azienda Ospedaliero Universitaria di Bologna, Italy; Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, S. Orsola-Malpighi Hospital, Italy
| | - Riccardo Casadei
- Division of Pancreatic Surgery, IRCCS, Azienda Ospedaliero Universitaria di Bologna, Italy; Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, S. Orsola-Malpighi Hospital, Italy
| |
Collapse
|
36
|
Nasti A, Inagaki S, Ho TTB, Seki A, Yoshida K, Satomura K, Sakai Y, Kaneko S, Yamashita T. Cystatin A promotes the antitumor activity of T helper type 1 cells and dendritic cells in murine models of pancreatic cancer. Mol Oncol 2025; 19:1452-1470. [PMID: 39792573 PMCID: PMC12077287 DOI: 10.1002/1878-0261.13796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 11/21/2024] [Accepted: 12/12/2024] [Indexed: 01/12/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a disease with poor prognosis due to diagnostic and therapeutic limitations. We previously identified cystatin A (CSTA) as a PDAC biomarker and have conducted the present study to investigate the antitumor effects of CSTA. PDAC murine models were established with genetically modified PAN02 tumor cell lines to evaluate the antitumor immune response. PDAC mouse survival was significantly longer with CSTA, and its antitumor effect was mediated mainly by CD4+ cells and partly by CD8+ cells. We also observed an increased infiltration of CD4+ and CD8+ cells in tumors of mice overexpressing CSTA. Phenotypically, we confirmed higher T helper type 1 (Th1) cell activity and increased frequency and activity of M1 macrophages and dendritic cells (DCs) in CSTA-overexpressing mice. Gene expression analysis highlighted pathways related to interferon gamma (IFN-γ) induction and Th1 lymphocyte activation that were induced by CSTA. Macrophages and DCs shifted toward proinflammatory antitumor phenotypes. Furthermore, activated splenocytes of PDAC model mice expressing CSTA had increased proapoptotic activity. CSTA also promoted the selective migration of CD4+ and CD11c+ immune cells in an in vitro migration assay. In conclusion, CSTA exerts antitumor effects by enhancing Th1-mediated antitumor effects through promotion of DC and M1 macrophage activity, thereby increasing immune cell chemotaxis. CSTA could be a novel therapeutic candidate for PDAC.
Collapse
Affiliation(s)
- Alessandro Nasti
- Information‐Based Medicine DevelopmentGraduate School of Medical Sciences, Kanazawa UniversityJapan
| | - Shingo Inagaki
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Tuyen Thuy Bich Ho
- Information‐Based Medicine DevelopmentGraduate School of Medical Sciences, Kanazawa UniversityJapan
| | - Akihiro Seki
- Department of GastroenterologyKanazawa University HospitalJapan
| | - Keiko Yoshida
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Kosuke Satomura
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Yoshio Sakai
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Shuichi Kaneko
- Information‐Based Medicine DevelopmentGraduate School of Medical Sciences, Kanazawa UniversityJapan
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
- Department of GastroenterologyKanazawa University HospitalJapan
| | - Taro Yamashita
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
- Department of GastroenterologyKanazawa University HospitalJapan
| |
Collapse
|
37
|
Rangelova E, Stoop TF, van Ramshorst TME, Ali M, van Bodegraven EA, Javed AA, Hashimoto D, Steyerberg E, Banerjee A, Jain A, Sauvanet A, Serrablo A, Giani A, Giardino A, Zerbi A, Arshad A, Wijma AG, Coratti A, Zironda A, Socratous A, Rojas A, Halimi A, Ejaz A, Oba A, Patel BY, Björnsson B, Reames BN, Tingstedt B, Goh BKP, Payá-Llorente C, Del Pozo CD, González-Abós C, Medin C, van Eijck CHJ, de Ponthaud C, Takishita C, Schwabl C, Månsson C, Ricci C, Thiels CA, Douchi D, Hughes DL, Kilburn D, Flanking D, Kleive D, Silva DS, Edil BH, Pando E, Moltzer E, Kauffman EF, Warren E, Bozkurt E, Sparrelid E, Thoma E, Verkolf E, Ausania F, Giannone F, Hüttner FJ, Burdio F, Souche FR, Berrevoet F, Daams F, Motoi F, Saliba G, Kazemier G, Roeyen G, Nappo G, Butturini G, Ferrari G, Kito Fusai G, Honda G, Sergeant G, Karteszi H, Takami H, Suto H, Matsumoto I, Mora-Oliver I, Frigerio I, Fabre JM, Chen J, Sham JG, Davide J, Urdzik J, de Martino J, Nielsen K, Okano K, Kamei K, Okada K, Tanaka K, Labori KJ, Goodsell KE, Alberici L, Webber L, Kirkov L, de Franco L, Miyashita M, Maglione M, Gramellini M, Ramera M, Amaral MJ, et alRangelova E, Stoop TF, van Ramshorst TME, Ali M, van Bodegraven EA, Javed AA, Hashimoto D, Steyerberg E, Banerjee A, Jain A, Sauvanet A, Serrablo A, Giani A, Giardino A, Zerbi A, Arshad A, Wijma AG, Coratti A, Zironda A, Socratous A, Rojas A, Halimi A, Ejaz A, Oba A, Patel BY, Björnsson B, Reames BN, Tingstedt B, Goh BKP, Payá-Llorente C, Del Pozo CD, González-Abós C, Medin C, van Eijck CHJ, de Ponthaud C, Takishita C, Schwabl C, Månsson C, Ricci C, Thiels CA, Douchi D, Hughes DL, Kilburn D, Flanking D, Kleive D, Silva DS, Edil BH, Pando E, Moltzer E, Kauffman EF, Warren E, Bozkurt E, Sparrelid E, Thoma E, Verkolf E, Ausania F, Giannone F, Hüttner FJ, Burdio F, Souche FR, Berrevoet F, Daams F, Motoi F, Saliba G, Kazemier G, Roeyen G, Nappo G, Butturini G, Ferrari G, Kito Fusai G, Honda G, Sergeant G, Karteszi H, Takami H, Suto H, Matsumoto I, Mora-Oliver I, Frigerio I, Fabre JM, Chen J, Sham JG, Davide J, Urdzik J, de Martino J, Nielsen K, Okano K, Kamei K, Okada K, Tanaka K, Labori KJ, Goodsell KE, Alberici L, Webber L, Kirkov L, de Franco L, Miyashita M, Maglione M, Gramellini M, Ramera M, Amaral MJ, Ramaekers M, Truty MJ, van Dam MA, Stommel MWJ, Petrikowski M, Imamura M, Hayashi M, D'Hondt M, Brunner M, Hogg ME, Zhang C, Suárez-Muñoz MÁ, Luyer MD, Unno M, Mizuma M, Janot M, Sahakyan MA, Jamieson NB, Busch OR, Bilge O, Belyaev O, Franklin O, Sánchez-Velázquez P, Pessaux P, Holka PS, Ghorbani P, Casadei R, Sartoris R, Schulick RD, Grützmann R, Sutcliffe R, Mata R, Patel RB, Takahashi R, Rodriguez Franco S, Cabús SS, Hirano S, Gaujoux S, Festen S, Kozono S, Maithel SK, Chai SM, Yamaki S, van Laarhoven S, Mieog JSD, Murakami T, Codjia T, Sumiyoshi T, Karsten TM, Nakamura T, Sugawara T, Boggi U, Hartman V, de Meijer VE, Bartholomä W, Kwon W, Koh YX, Cho Y, Takeyama Y, Inoue Y, Nagakawa Y, Kawamoto Y, Ome Y, Soonawalla Z, Uemura K, Wolfgang CL, Jang JY, Padbury R, Satoi S, Messersmith W, Wilmink JW, Abu Hilal M, Besselink MG, Del Chiaro M. The impact of neoadjuvant therapy in patients with left-sided resectable pancreatic cancer: an international multicenter study. Ann Oncol 2025; 36:529-542. [PMID: 39814200 DOI: 10.1016/j.annonc.2024.12.015] [Show More Authors] [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: 09/04/2024] [Revised: 10/26/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Left-sided pancreatic cancer is associated with worse overall survival (OS) compared with right-sided pancreatic cancer. Although neoadjuvant therapy is currently seen as not effective in patients with resectable pancreatic cancer (RPC), current randomized trials included mostly patients with right-sided RPC. The purpose of this study was to assess the association between neoadjuvant therapy and OS in patients with left-sided RPC compared with upfront surgery. PATIENTS AND METHODS This was an international multicenter retrospective study including consecutive patients after left-sided pancreatic resection for pathology-proven RPC, either after neoadjuvant therapy or upfront surgery in 76 centers from 18 countries on 4 continents (2013-2019). The primary endpoint was OS from diagnosis. Time-dependent Cox regression analysis was carried out to investigate the association of neoadjuvant therapy with OS, adjusting for confounders at the time of diagnosis. Adjusted OS probabilities were calculated. RESULTS Overall, 2282 patients after left-sided pancreatic resection for RPC were included of whom 290 patients (13%) received neoadjuvant therapy. The most common neoadjuvant regimens were (m)FOLFIRINOX (38%) and gemcitabine-nab-paclitaxel (22%). After upfront surgery, 72% of patients received adjuvant chemotherapy, mostly a single-agent regimen (74%). Neoadjuvant therapy was associated with prolonged OS compared with upfront surgery (adjusted hazard ratio 0.69, 95% confidence interval 0.58-0.83) with an adjusted median OS of 53 versus 37 months (P = 0.0003) and adjusted 5-year OS rates of 47% versus 35% (P = 0.0001) compared with upfront surgery. Interaction analysis demonstrated a stronger effect of neoadjuvant therapy in patients with a larger tumor (Pinteraction = 0.003) and higher serum carbohydrate antigen 19-9 (CA19-9; Pinteraction = 0.005). In contrast, the effect of neoadjuvant therapy was not enhanced for splenic artery (Pinteraction = 0.43), splenic vein (Pinteraction = 0.30), retroperitoneal (Pinteraction = 0.84), and multivisceral (Pinteraction = 0.96) involvement. CONCLUSIONS Neoadjuvant therapy in patients with left-sided RPC was associated with improved OS compared with upfront surgery. The impact of neoadjuvant therapy increased with larger tumor size and higher serum CA19-9 at diagnosis. Randomized controlled trials on neoadjuvant therapy specifically in patients with left-sided RPC are needed.
Collapse
Affiliation(s)
- E Rangelova
- Department of Upper Abdominal Surgery at Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - T F Stoop
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands; Cancer Center Amsterdam, Amsterdam, The Netherlands; Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, USA
| | - T M E van Ramshorst
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands; Cancer Center Amsterdam, Amsterdam, The Netherlands; Department of Surgery, Fondazione Poliambulanza, Instituto Ospedaliero, Brescia, Italy
| | - M Ali
- Cancer Center Amsterdam, Amsterdam, The Netherlands; Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, The Netherlands
| | - E A van Bodegraven
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands; Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - A A Javed
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands; Cancer Center Amsterdam, Amsterdam, The Netherlands; Department of Surgical Oncology, Department of Surgery, New York University Medical Center, New York, USA
| | - D Hashimoto
- Department of Surgery, Kansai Medical University, Osaka, Japan
| | - E Steyerberg
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - A Banerjee
- HPB & Liver Transplant Unit, Royal Free Hospital, London, UK
| | - A Jain
- Division of Surgical Oncology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - A Sauvanet
- Department of HPB Surgery and Liver Transplantation, APHP Beaujon Hospital, University of Paris Cité, Clichy, France
| | - A Serrablo
- HPB Surgical Division, Miguel Servet University Hospital, Zaragoza, Spain
| | - A Giani
- Division of Minimally-Invasive Surgical Oncology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - A Giardino
- Department of HPB Surgery, Pederzoli Hospital, Peschiera del Garda, Peschiera, Italy
| | - A Zerbi
- Humanitas University, Department of Biomedical Sciences, Milan, Italy; Pancreatic Surgery Unit, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| | - A Arshad
- Hepatopancreatobiliary Unit, University Hospitals Southampton NHS Trust, Southampton General Hospital, Southampton, UK
| | - A G Wijma
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - A Coratti
- General and Emergency Surgery Unit, Misericordia Hospital, Azienda USL Toscana Sud-Est, Grosseto, Italy
| | - A Zironda
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, USA
| | - A Socratous
- Department of Upper Abdominal Surgery at Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - A Rojas
- Department of Surgery, NorthShore University Health System, Evanston, USA
| | - A Halimi
- Department of Surgery, Umeå University, Umeå, Sweden; Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet at Karolinska University Hospital, Stockholm, Sweden
| | - A Ejaz
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, USA
| | - A Oba
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, USA; Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo; Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | - B Y Patel
- Hepatopancreatobiliary Unit, University Hospitals Southampton NHS Trust, Southampton General Hospital, Southampton, UK
| | - B Björnsson
- Department of Surgery, Linköping University Hospital, Linköping, Sweden
| | - B N Reames
- Department of Surgery, University of Nebraska Medical Center, Omaha, USA
| | - B Tingstedt
- Department of Clinical Sciences Lund, Surgery, Lund University, Skåne University Hospital, Lund, Sweden
| | - B K P Goh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore, Singapore
| | - C Payá-Llorente
- General and Digestive Surgery, Hospital Doctor Peset, Valencia, Spain
| | - C D Del Pozo
- General and Digestive Surgery, Hospital Doctor Peset, Valencia, Spain
| | - C González-Abós
- Hepatobiliopancreatic Department, Hospital Clinic de Barcelona, Barcelona, Spain
| | - C Medin
- Division of Surgical Oncology, Winship Cancer Institute, Emory University, Atlanta, USA
| | - C H J van Eijck
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - C de Ponthaud
- Department of Hepatobiliary and Pancreatic Surgery and Liver Transplantation, AP-HP, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - C Takishita
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, Tokyo, Japan
| | - C Schwabl
- Department of Radiology, Medical University Innsbruck, Innsbruck, Austria
| | - C Månsson
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - C Ricci
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, Bologna, Italy; Division of Pancreatic Surgery, IRCCS, Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - C A Thiels
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, USA
| | - D Douchi
- Department of Surgery, Tohoku University, Sendai, Japan
| | - D L Hughes
- Department of Hepatobiliary and Pancreatic Surgery, Oxford Radcliffe Hospitals NHS Foundation Trust, Oxford, UK
| | - D Kilburn
- Department of Surgery, Flinders Medical Centre, Adelaide, Australia
| | - D Flanking
- Department of Upper Abdominal Surgery at Sahlgrenska University Hospital, Gothenburg, Sweden
| | - D Kleive
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Oslo, Norway
| | - D S Silva
- HEBIPA Surgery, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - B H Edil
- Division of Surgical Oncology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - E Pando
- Universitat Autónoma de Barcelona, Barcelona, Spain; Department of Hepato-Pancreato-Biliary and Transplant Surgery, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - E Moltzer
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - E F Kauffman
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | - E Warren
- Division of Surgical Oncology, Winship Cancer Institute, Emory University, Atlanta, USA
| | - E Bozkurt
- Department of General Surgery, Koç University School of Medicine, Istanbul, Turkey
| | - E Sparrelid
- Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet at Karolinska University Hospital, Stockholm, Sweden
| | - E Thoma
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| | - E Verkolf
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - F Ausania
- Hepatobiliopancreatic Department, Hospital Clinic de Barcelona, Barcelona, Spain; Instituto de Investigaciones Biomédicas August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - F Giannone
- Department of Visceral and Digestive Surgery, University Hospital of Strasbourg, Strasbourg, France
| | - F J Hüttner
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| | - F Burdio
- Department of Surgery, Hepatobiliary and Pancreatic Unit, Hospital del Mar de Barcelona, Barcelona, Spain; Hospital del Mar Research Institute (IMIM), University Pompeu-Fabra, Barcelona, Spain
| | - F R Souche
- Oncologic & Minimally-Invasive Digestive Surgery, CHU Montpellier, University of Montpellier, Montpellier, France
| | - F Berrevoet
- Department of General and HPB Surgery and Liver Transplantation, Ghent University Hospital, Ghent, Belgium
| | - F Daams
- Cancer Center Amsterdam, Amsterdam, The Netherlands; Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, The Netherlands
| | - F Motoi
- Department of Surgery, Yamagata University, Yamagata, Japan
| | - G Saliba
- Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet at Karolinska University Hospital, Stockholm, Sweden
| | - G Kazemier
- Cancer Center Amsterdam, Amsterdam, The Netherlands; Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, The Netherlands
| | - G Roeyen
- Department of HPB, Endocrine and Transplantation Surgery, University Hospital Antwerp, Antwerp, Belgium
| | - G Nappo
- Humanitas University, Department of Biomedical Sciences, Milan, Italy; Pancreatic Surgery Unit, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| | - G Butturini
- Department of HPB Surgery, Pederzoli Hospital, Peschiera del Garda, Peschiera, Italy
| | - G Ferrari
- Division of Minimally-Invasive Surgical Oncology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - G Kito Fusai
- HPB & Liver Transplant Unit, Royal Free Hospital, London, UK
| | - G Honda
- Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - G Sergeant
- Department of Abdominal Surgery, Jessa Hospital, Faculty of Medicine and Health Sciences, Universiteit Hasselt, Hasselt, Belgium
| | - H Karteszi
- Department of Radiology, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - H Takami
- Department of Gastroenterological Surgery (Surgery II), Nagoya University, Nagoya, Japan
| | - H Suto
- Department of Gastroenterological Surgery, Kagawa University, Kagawa, Japan
| | - I Matsumoto
- Department of Surgery, Kindai University, Osakasayama, Japan
| | - I Mora-Oliver
- Department of Surgery, Liver and Pancreato-Biliary Unit, Hospital Clínico Universitario Valencia, Biomedical Research Institute, INCLIVA, Valencia, Spain
| | - I Frigerio
- Department of HPB Surgery, Pederzoli Hospital, Peschiera del Garda, Peschiera, Italy
| | - J M Fabre
- Oncologic & Minimally-Invasive Digestive Surgery, CHU Montpellier, University of Montpellier, Montpellier, France
| | - J Chen
- Department of Surgery, Flinders Medical Centre, Adelaide, Australia
| | - J G Sham
- Department of Surgery, University of Washington, Seattle, USA; Fred Hutchinson Cancer Center, Seattle, USA
| | - J Davide
- HEBIPA Surgery, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - J Urdzik
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - J de Martino
- Department of Hepatobiliary and Pancreatic Surgery and Liver Transplantation, AP-HP, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - K Nielsen
- Department of Hepatopancreatobiliary Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - K Okano
- Department of Gastroenterological Surgery, Kagawa University, Kagawa, Japan
| | - K Kamei
- Department of Surgery, Kindai University, Osakasayama, Japan
| | - K Okada
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - K Tanaka
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Hokkaido, Japan
| | - K J Labori
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - K E Goodsell
- Department of Surgery, University of Washington, Seattle, USA
| | - L Alberici
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - L Webber
- Department of Upper GI Surgery, Fiona Stanley Hospital, Perth, Austria
| | - L Kirkov
- Oncologic & Minimally-Invasive Digestive Surgery, CHU Montpellier, University of Montpellier, Montpellier, France
| | - L de Franco
- General and Emergency Surgery Unit, Misericordia Hospital, Azienda USL Toscana Sud-Est, Grosseto, Italy
| | - M Miyashita
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | - M Maglione
- Department of Visceral, Transplant, and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - M Gramellini
- Humanitas University, Department of Biomedical Sciences, Milan, Italy; Pancreatic Surgery Unit, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| | - M Ramera
- Department of Surgery, Fondazione Poliambulanza, Instituto Ospedaliero, Brescia, Italy; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - M J Amaral
- General Surgery Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - M Ramaekers
- Department of Surgery, Catharina Hospital, Eindhoven, The Netherlands
| | - M J Truty
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, USA
| | - M A van Dam
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - M W J Stommel
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M Petrikowski
- Department of Surgery, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - M Imamura
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University, Hokkaido, Japan
| | - M Hayashi
- Department of Gastroenterological Surgery (Surgery II), Nagoya University, Nagoya, Japan
| | - M D'Hondt
- Department of Digestive and Hepatobiliary-Pancreatic Surgery, Groeninge Hospital, Kortrijk, Belgium
| | - M Brunner
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen Nürnberg, Erlangen, Germany
| | - M E Hogg
- Department of Surgery, NorthShore University Health System, Evanston, USA
| | - C Zhang
- Department of Surgery, University of Nebraska Medical Center, Omaha, USA
| | - M Á Suárez-Muñoz
- HPB Surgical Unit, University Hospital Virgen de la Victoria, Málaga, Spain
| | - M D Luyer
- Department of Surgery, Catharina Hospital, Eindhoven, The Netherlands
| | - M Unno
- Department of Surgery, Tohoku University, Sendai, Japan
| | - M Mizuma
- Department of Surgery, Tohoku University, Sendai, Japan
| | - M Janot
- Department of Surgery, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - M A Sahakyan
- The Intervention Center, Oslo University Hospital, Rigshospitalet, Oslo, Norway
| | - N B Jamieson
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - O R Busch
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands; Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - O Bilge
- Department of General Surgery, Koç University School of Medicine, Istanbul, Turkey
| | - O Belyaev
- Department of Surgery, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - O Franklin
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, USA; Department of Surgery, Umeå University, Umeå, Sweden
| | - P Sánchez-Velázquez
- Department of Surgery, Hepatobiliary and Pancreatic Unit, Hospital del Mar de Barcelona, Barcelona, Spain; Hospital del Mar Research Institute (IMIM), University Pompeu-Fabra, Barcelona, Spain
| | - P Pessaux
- Department of Visceral and Digestive Surgery, University Hospital of Strasbourg, Strasbourg, France
| | - P S Holka
- Department of Clinical Sciences Lund, Surgery, Lund University, Skåne University Hospital, Lund, Sweden
| | - P Ghorbani
- Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet at Karolinska University Hospital, Stockholm, Sweden
| | - R Casadei
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, Bologna, Italy; Division of Pancreatic Surgery, IRCCS, Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - R Sartoris
- Department of Radiology, APHP Beaujon Hospital, University of Paris Cité, Clichy, France
| | - R D Schulick
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, USA
| | - R Grützmann
- The Intervention Center, Oslo University Hospital, Rigshospitalet, Oslo, Norway
| | - R Sutcliffe
- Department of Hepatopancreatobiliary Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - R Mata
- Universitat Autónoma de Barcelona, Barcelona, Spain; Department of Hepato-Pancreato-Biliary and Transplant Surgery, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - R B Patel
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, USA
| | - R Takahashi
- Department of Surgery, Yamagata University, Yamagata, Japan
| | - S Rodriguez Franco
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, USA
| | - S S Cabús
- Department of HPB Surgery, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - S Hirano
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Hokkaido, Japan
| | - S Gaujoux
- Department of Hepatobiliary and Pancreatic Surgery and Liver Transplantation, AP-HP, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - S Festen
- Department of Surgery, OLVG, Amsterdam, The Netherlands
| | - S Kozono
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, Tokyo, Japan
| | - S K Maithel
- Division of Surgical Oncology, Winship Cancer Institute, Emory University, Atlanta, USA
| | - S M Chai
- Department of Anatomical Pathology, PathWest Laboratory Medicine, Perth, Australia
| | - S Yamaki
- Department of Surgery, Kansai Medical University, Osaka, Japan
| | - S van Laarhoven
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands; Department of HPB Surgery, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - J S D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - T Murakami
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University, Hokkaido, Japan
| | - T Codjia
- Department of Digestive Surgery, Rouen University Hospital, Rouen, France
| | - T Sumiyoshi
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - T M Karsten
- Department of Surgery, OLVG, Amsterdam, The Netherlands
| | - T Nakamura
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Hokkaido, Japan
| | - T Sugawara
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, USA; Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo
| | - U Boggi
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | - V Hartman
- Department of HPB, Endocrine and Transplantation Surgery, University Hospital Antwerp, Antwerp, Belgium
| | - V E de Meijer
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - W Bartholomä
- Department of Radiology, Linköping University, Linköping, Sweden
| | - W Kwon
- Department of Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Y X Koh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore, Singapore
| | - Y Cho
- Department of Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Y Takeyama
- Department of Surgery, Kindai University, Osakasayama, Japan
| | - Y Inoue
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan
| | - Y Nagakawa
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, Tokyo, Japan
| | - Y Kawamoto
- Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - Y Ome
- Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - Z Soonawalla
- Department of Hepatobiliary and Pancreatic Surgery, Oxford Radcliffe Hospitals NHS Foundation Trust, Oxford, UK
| | - K Uemura
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - C L Wolfgang
- Department of Surgical Oncology, Department of Surgery, New York University Medical Center, New York, USA
| | - J Y Jang
- Department of Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - R Padbury
- Department of Surgery, Flinders Medical Centre, Adelaide, Australia
| | - S Satoi
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, USA; Department of Surgery, Kansai Medical University, Osaka, Japan
| | - W Messersmith
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, USA
| | - J W Wilmink
- Cancer Center Amsterdam, Amsterdam, The Netherlands; Amsterdam UMC, Location University of Amsterdam, Department of Medical Oncology, Amsterdam, The Netherlands
| | - M Abu Hilal
- Department of HPB Surgery, Pederzoli Hospital, Peschiera del Garda, Peschiera, Italy
| | - M G Besselink
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands; Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - M Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, USA
| |
Collapse
|
38
|
Hays SB, Kuchta K, Abreu AA, Chopra A, Farah E, Kannan A, Mehdi SA, Radi I, Ranson K, Rieser C, Rojas AE, Tcharni A, Boone BA, Paniccia A, Polanco PM, Schmidt CR, Talamonti MS, Zeh HJ, Zureikat AH, Hogg ME. Multimodal Therapy May be Key to Improving Survival for Octogenarians Undergoing Pancreaticoduodenectomy for Pancreatic Ductal Adenocarcinoma: An American Multicenter Analysis. Ann Surg Oncol 2025; 32:3582-3592. [PMID: 39871074 DOI: 10.1245/s10434-025-16916-7] [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: 09/25/2024] [Accepted: 01/07/2025] [Indexed: 01/29/2025]
Abstract
BACKGROUND As the population ages, the number of octogenarians with pancreatic ductal adenocarcinoma (PDAC) continues to rise. Morbidity and mortality following pancreatectomy have improved owing to safer surgery and better chemoradiation regimens. This study compares the outcomes and multimodality utilization in octogenarians (≥80 years) who underwent pancreaticoduodenectomy (PD) for PDAC, with a younger cohort. METHODS This is a multi-institutional retrospective review from 2007 to 2023 of patients who underwent PD for PDAC. Preoperative, perioperative, and oncologic outcomes were analyzed, and multivariable analysis (MVA) was performed. RESULTS A total of 1,051 patients underwent PD for PDAC ( ≥ 80 = 125, < 80 = 926). Octogenarians had a higher age unadjusted Charlson Comorbidity Index (p = 0.0146) and were more likely to have prior abdominal surgery (p = 0.0019). Patients <80 years received chemotherapy (p < 0.0001) or radiation (p < 0.0001), including neoadjuvant chemotherapy (p < 0.0001), more frequently than octogenarians, who more commonly underwent upfront surgery (p < 0.0001). There were no significant differences in complications. Octogenarians had a higher 90 day mortality rate (7.2% versus 3.5%, p = 0.0424); however, this was not significant on MVA. The <80 cohort had longer overall survival (OS) (p = 0.0004). Receiving any chemotherapy was associated with longer OS (hazard ratio [HR] 0.59 [0.46-0.75], p < 0.0001). In survival analysis of octogenarians only, receiving multimodal therapy significantly prolonged OS compared with surgery alone (p = 0.0349). CONCLUSIONS Octogenarian status does not increase morbidity or mortality but is associated with decreased survival in PDAC patients undergoing PD. Chemotherapy had a protective effect on OS; however, octogenarians received less multimodal therapy compared with the younger cohort.
Collapse
Affiliation(s)
- Sarah B Hays
- Department of Surgery, NorthShore University Health System, Evanston, IL, USA.
- Department of Surgery, University of Chicago, Chicago, IL, USA.
| | - Kristine Kuchta
- Department of Surgery, NorthShore University Health System, Evanston, IL, USA
| | - Andres A Abreu
- Department of Surgery, University of Texas at Southwestern, Dallas, TX, USA
| | - Asmita Chopra
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Emile Farah
- Department of Surgery, University of Texas at Southwestern, Dallas, TX, USA
| | - Amudhan Kannan
- Department of Surgery, University of Texas at Southwestern, Dallas, TX, USA
| | - Syed Abbas Mehdi
- Department of Surgery, NorthShore University Health System, Evanston, IL, USA
| | - Imad Radi
- Department of Surgery, University of Texas at Southwestern, Dallas, TX, USA
| | - Kristen Ranson
- Department of Surgery, West Virginia University, Morgantown, WV, USA
| | - Caroline Rieser
- Department of Surgery, NorthShore University Health System, Evanston, IL, USA
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Aram E Rojas
- Department of Surgery, NorthShore University Health System, Evanston, IL, USA
| | - Adam Tcharni
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Brian A Boone
- Department of Surgery, West Virginia University, Morgantown, WV, USA
| | - Alessandro Paniccia
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Patricio M Polanco
- Department of Surgery, University of Texas at Southwestern, Dallas, TX, USA
| | - Carl R Schmidt
- Department of Surgery, West Virginia University, Morgantown, WV, USA
| | - Mark S Talamonti
- Department of Surgery, NorthShore University Health System, Evanston, IL, USA
| | - Herbert J Zeh
- Department of Surgery, University of Texas at Southwestern, Dallas, TX, USA
| | - Amer H Zureikat
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Melissa E Hogg
- Department of Surgery, NorthShore University Health System, Evanston, IL, USA
| |
Collapse
|
39
|
Jacobsen A, Flessa M, Abels AL, Czubayko F, Mittelstädt A, Krautz C, Weber GF, Grützmann R, Brunner M. Impact of Structured Postoperative Surveillance on Survival in Patients with Resected Pancreatic Adenocarcinoma. Cancers (Basel) 2025; 17:1424. [PMID: 40361350 PMCID: PMC12070849 DOI: 10.3390/cancers17091424] [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: 03/08/2025] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND This study aimed to investigate whether structured surveillance after resection of PDAC has an impact on survival. METHODS 206 patients who underwent PDAC resection at the University Hospital Erlangen between 2005 and 2020 and survived for at least 90 days postoperatively were included in this retrospective study. The impact of surveillance (structured vs. no structured surveillance) respectively of recurrence patterns (recurrence in follow-up vs. symptomatic recurrence) on overall and disease-free survival was investigated. RESULTS A total of 157 patients (76%) participated in a structured surveillance program after pancreatic resection. During a mean follow-up of 28.5 months, 137 patients (67%) developed recurrence. Patients with structured surveillance showed significantly better outcomes for overall survival after surgery (OSaS: 29.2 months with structured surveillance vs. 16.4 months without, p < 0.001), overall survival after recurrence (OSaR: 10.8 months vs. 3.6 months, p < 0.001), and disease-free survival (DFS: 14.8 months vs. 11.4 months, p = 0.010). In a subgroup analysis of 112 patients with recurrence, survival benefits were observed for those whose recurrence was detected during follow-up compared to those with symptomatic recurrence (OSaS: 24.8 months in the follow-up group vs. 17.2 months in the symptomatic group, p < 0.001; OSaR: 12.6 months vs. 6.5 months, p < 0.001). CONCLUSION This study provides evidence that structured surveillance after PDAC resection is beneficial for both overall and disease-free survival. However, randomized studies are needed to confirm the positive impact of structured surveillance programs on survival after pancreatic resection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Maximilian Brunner
- Department of General and Visceral Surgery, University Hospital Erlangen, Friedrich-Alexander-University (FAU), 91054 Erlangen, Germany; (A.J.); (M.F.); (A.-L.A.); (F.C.); (A.M.); (C.K.); (G.F.W.); (R.G.)
| |
Collapse
|
40
|
Okada Y, Sato Y, Shinomiya R, Miyake T, Takahashi T, Yokoyama R, Mitsui Y, Tomonari T, Okamoto K, Sogabe M, Miyamoto H, Kawano Y, Takayama T. Conditions for effective use of liposomal irinotecan with fluorouracil and leucovorin in unresectable pancreatic cancer after FOLFIRINOX treatment. Int J Clin Oncol 2025:10.1007/s10147-024-02677-y. [PMID: 40266453 DOI: 10.1007/s10147-024-02677-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/01/2024] [Indexed: 04/24/2025]
Abstract
BACKGROUND Liposomal irinotecan + fluorouracil/leucovorin (nal-IRI + 5FU/LV) is commonly used as a second- or later-line treatment for pancreatic ductal adenocarcinoma (PDAC) and offers survival benefits. However, its efficacy and safety in patients previously treated with FOLFIRINOX, which includes irinotecan, remain unclear. We evaluated the efficacy and safety of nal-IRI + 5FU/LV in patients with unresectable PDAC who received previous FOLFIRINOX therapy and those who did not. METHODS This retrospective observational study included 42 patients with PDAC who were treated with nal-IRI + 5FU/LV (October 2020-November 2023). Patients were grouped based on prior FOLFIRINOX treatment. RESULTS The progression-free survival (PFS) in patients who previously received modified FOLFIRINOX (mFFX) therapy was shorter than that in patients who did not (2.5 vs. 3.5 months, P = 0.07). When patients with greater than- and less than the cut-off value of irinotecan-free interval (IFI) were classified into the long and short IFI groups, respectively, PFS was significantly longer in the long-IFI group than that in the short IFI group (4.0 vs. 2.1 months, P = 0.01). Moreover, the C-reactive protein/albumin ratio (CAR) was also a significant predictor of PFS (P = 0.03). Furthermore, both factors were found to be independent factors influencing PFS in the univariate Cox regression analysis (P = 0.02 and P = 0.04). CONCLUSION Nal-IRI + 5FU/LV therapy may be a safe and effective option as a second- or later-line treatment, particularly for patients who have not previously received mFFX therapy. For patients who received prior mFFX exposure, a longer IFI and lower CAR may indicate greater potential benefit, thus aiding in more personalized treatment approaches.
Collapse
Affiliation(s)
- Yasuyuki Okada
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Yasushi Sato
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan.
- Department of Community Medicine for Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Ryo Shinomiya
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Takanori Miyake
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Taku Takahashi
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Reiko Yokoyama
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Yasuhiro Mitsui
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Tetsu Tomonari
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Koichi Okamoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Masahiro Sogabe
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Hiroshi Miyamoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Yutaka Kawano
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
41
|
Xia J, Zheng L, Zhang H, Fan Q, Liu H, Wang O, Yan H. Drug Resistance Analysis of Pancreatic Cancer Based on Universally Differentially Expressed Genes. Int J Mol Sci 2025; 26:3936. [PMID: 40362181 PMCID: PMC12071644 DOI: 10.3390/ijms26093936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
The high heterogeneity between patients can complicate the diagnosis and treatment of pancreatic ductal adenocarcinoma (PDAC). Here, we explored the association of universally differentially expressed genes (UDEGs) with resistance to chemotherapy and immunotherapy in the context of pancreatic cancer. In this work, sixteen up-regulated and three down-regulated genes that were dysregulated in more than 85% of 102 paired and 5% of 521 unpaired PDAC samples were identified and defined as UDEGs. A single-cell level analysis further validated the high expression levels of the up-UDEGs and the low levels of the down-UDEGs in cancer-related ductal cells, which could represent the malignant changes seen in pancreatic cancer. Based on a drug sensitivity analysis, we found that ANLN, GPRC5A and SERPINB5 are closely related to the resistance mechanism of PDAC, and their high expression predicted worse survival for PDAC patients. This suggests that targeting these genes could be a potential way to reduce drug resistance and improve survival. Based on the immune infiltration analysis, the abnormal expression of the UDEGs was found to be related to the formation of an immunosuppressive tumor microenvironment. In conclusion, these UDEGs are common features of PDAC and could be involved in the resistance of pancreatic cancer and might serve as novel drug targets to guide research into drug repurposing.
Collapse
Affiliation(s)
- Jie Xia
- School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, China;
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China; (L.Z.); (H.Z.); (Q.F.); (H.L.); (O.W.)
| | - Linyong Zheng
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China; (L.Z.); (H.Z.); (Q.F.); (H.L.); (O.W.)
| | - Huarong Zhang
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China; (L.Z.); (H.Z.); (Q.F.); (H.L.); (O.W.)
| | - Qi Fan
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China; (L.Z.); (H.Z.); (Q.F.); (H.L.); (O.W.)
| | - Hui Liu
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China; (L.Z.); (H.Z.); (Q.F.); (H.L.); (O.W.)
| | - Ouxi Wang
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China; (L.Z.); (H.Z.); (Q.F.); (H.L.); (O.W.)
| | - Haidan Yan
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China; (L.Z.); (H.Z.); (Q.F.); (H.L.); (O.W.)
| |
Collapse
|
42
|
Li L, Wu ZT, Duan WH, Liu J, Zhu YR. Machine learning-based prognostic modelling of NK cells in PAAD for immunotherapy guidance. Discov Oncol 2025; 16:577. [PMID: 40253675 PMCID: PMC12009793 DOI: 10.1007/s12672-025-02266-z] [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: 01/18/2025] [Accepted: 03/28/2025] [Indexed: 04/22/2025] Open
Abstract
Pancreatic cancer's high incidence and mortality rates are underscored by ineffective treatments, particularly immunotherapy's poor performance. This could stem from an unclear immune microenvironment, where NK cells may play a unique role. Analyzing the NK cell-differentially expressed genes (NKDEGs) from the PAAD_GSE162708 single-cell dataset and utilizing the TCGA-PAAD and ICGC-PACA-AU datasets, we identified 11 NKDEGs linked to pancreatic adenocarcinoma (PAAD) prognosis and developed a prognostic model. This model's risk scores significantly outperformed traditional grading and TNM staging systems, validated through clinical and pathological analyses. Functional enrichment analysis pointed to the Neuroactive ligand-receptor interaction and MAPK signaling pathways, suggesting NK cells' distinctive role in PAAD. High-risk groups showed decreased overall NK cells but increased activated NK cells, which may mediate adverse inflammatory responses. NK cells exhibit synergistic interactions with plasma cells and macrophages and negative regulation by monocytes and naive B cells. Our model accurately predicts immunotherapy responses, indicating potential for targeted drugs to enhance treatment. Additionally, we introduced an NKDEGs-based immunotyping approach for personalized medicine and clinical decision-making in PAAD. This study emphasizes NK cells' potential in PAAD treatment, offering precise patient stratification and therapeutic targets for immunotherapy.
Collapse
Affiliation(s)
- Li Li
- The People'S Hospital of Wenshan Prefecture, Kunming University of Science and Technology, No. 228, Kaihua East Road, Wenshan, 663000, Yunnan, China
| | - Zu-Tao Wu
- The People'S Hospital of Wenshan Prefecture, Kunming University of Science and Technology, No. 228, Kaihua East Road, Wenshan, 663000, Yunnan, China
| | - Wen-Hong Duan
- The People'S Hospital of Wenshan Prefecture, Kunming University of Science and Technology, No. 228, Kaihua East Road, Wenshan, 663000, Yunnan, China
| | - Jiang Liu
- The People'S Hospital of Wenshan Prefecture, Kunming University of Science and Technology, No. 228, Kaihua East Road, Wenshan, 663000, Yunnan, China
| | - Yin-Rong Zhu
- The People'S Hospital of Wenshan Prefecture, Kunming University of Science and Technology, No. 228, Kaihua East Road, Wenshan, 663000, Yunnan, China.
| |
Collapse
|
43
|
Yeo D, Seyfi D, Bastian A, Strauss H, Leach A, Klemm V, Pirrello A, Spring K, Saxena P, Wahlroos S, Sutherland S, Grimison P, Park JS, Sandroussi C, Rasko JE. Portal venous circulating tumor cells as a biomarker for relapse prediction in resected pancreatic cancer. Cell Mol Life Sci 2025; 82:155. [PMID: 40208273 PMCID: PMC11985722 DOI: 10.1007/s00018-025-05669-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: 11/11/2024] [Revised: 03/16/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Pancreatic cancer is an aggressive disease with poor prognosis. The only potentially curative treatment option is surgical resection, however recurrence is common. Biomarkers to detect minimal residual disease, assist with risk stratification, relapse and real time monitoring, are required. Circulating tumor cells (CTCs) are a promising liquid biopsy biomarker for solid tumors. However, their role in monitoring minimal residual disease in pancreatic cancer remains to be determined. Our study aimed to investigate whether detection and enumeration of CTCs could predict recurrence and provide monitoring of disease status. METHOD Participants planned for Whipple procedure or partial pancreatectomy were enrolled in this prospective pilot study. Intraoperatively, 7.5 mL of portal and peripheral venous blood were collected, and peripheral venous blood was also collected post-surgery. CTC identification and enumeration were performed using the AccuCyte-CyteFinder platform and CellSieve microfiltration. RESULTS Of 29 participants, 20 were confirmed to have epithelial cancer by histopathology, where 15 had pancreatic ductal adenocarcinoma. In those with epithelial cancer, CTCs were detected intraoperatively in 75% of portal venous blood samples, in contrast to 40% detected in peripheral venous blood (median: 6 and 0 per 7.5mL respectively). Only portal venous CTC detection was predictive of pancreatic ductal adenocarcinoma relapse. The positive (> 5) portal venous CTC group had a 6.67 times higher risk of recurring (odds ratio = 20.43, sensitivity = 1.00, specificity = 0.625). Detection of peripheral venous CTCs post-surgery was also correlated with relapse in a small subset of patients. CONCLUSIONS If validated, CTCs may provide a prognostic and monitoring biomarker in patients with pancreatic cancer undergoing surgery.
Collapse
Affiliation(s)
- Dannel Yeo
- Li Ka Shing Cell & Gene Therapy Program, The University of Sydney, Camperdown, Australia
- Precision Oncology Laboratory, Cancer Innovations, Centenary Institute, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| | - Doruk Seyfi
- Department of Hepatobiliary and Upper Gastrointestinal Surgery, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
- Upper Gastrointestinal Surgery, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Althea Bastian
- Li Ka Shing Cell & Gene Therapy Program, The University of Sydney, Camperdown, Australia
- Precision Oncology Laboratory, Cancer Innovations, Centenary Institute, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| | - Heidi Strauss
- Li Ka Shing Cell & Gene Therapy Program, The University of Sydney, Camperdown, Australia
- Precision Oncology Laboratory, Cancer Innovations, Centenary Institute, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| | - Anna Leach
- Li Ka Shing Cell & Gene Therapy Program, The University of Sydney, Camperdown, Australia
- Precision Oncology Laboratory, Cancer Innovations, Centenary Institute, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| | - Vera Klemm
- Li Ka Shing Cell & Gene Therapy Program, The University of Sydney, Camperdown, Australia
- Precision Oncology Laboratory, Cancer Innovations, Centenary Institute, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| | - Anthony Pirrello
- Li Ka Shing Cell & Gene Therapy Program, The University of Sydney, Camperdown, Australia
- Precision Oncology Laboratory, Cancer Innovations, Centenary Institute, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Kevin Spring
- Medical Oncology Group, Liverpool Clinical School, School of Medicine, Western Sydney University and Ingham Institute for Applied Medical Research, Liverpool, Australia
- Concord Institute of Academic Surgery, Sydney Local Health District, Concord, Australia
- South-West Sydney Clinical Campuses, UNSW Medicine & Health, Sydney, Australia
| | - Payal Saxena
- Division of Gastroenterology, Department of Medicine, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
- Gastroenterology, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Sara Wahlroos
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Sarah Sutherland
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Peter Grimison
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Jin-Soo Park
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Department of Hepatobiliary and Upper Gastrointestinal Surgery, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
- Upper Gastrointestinal Surgery, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Charbel Sandroussi
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Department of Hepatobiliary and Upper Gastrointestinal Surgery, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
- Upper Gastrointestinal Surgery, Chris O'Brien Lifehouse, Camperdown, Australia
| | - John Ej Rasko
- Li Ka Shing Cell & Gene Therapy Program, The University of Sydney, Camperdown, Australia.
- Precision Oncology Laboratory, Cancer Innovations, Centenary Institute, Camperdown, Australia.
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia.
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia.
| |
Collapse
|
44
|
Manne A, Bao Y, Sheel A, Sara A, Manne U, Thanikachalam K, Esnakula A, Pawlik TM, Cloyd JM, Tsai S, Kasi A, Paluri RK, Sherpally D, Jeepalyam S, Yu L, Yang W. Prognostic significance of serum MUC5AC in resected pancreatic ductal adenocarcinoma: initial insights. Front Oncol 2025; 15:1544928. [PMID: 40260290 PMCID: PMC12010103 DOI: 10.3389/fonc.2025.1544928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
Background We investigated the association between serum MUC5AC (sMUC5AC) levels and patient outcomes in individuals who underwent resection for pancreatic ductal adenocarcinoma (PDA), including those treated with neoadjuvant therapy (NAT) and those who had upfront surgery (UpS) followed by adjuvant therapy. Methods Serum samples from the Ohio State University biorepository collected from January 2010 to June 2021 were utilized. The human MUC5AC kit (NBP2-76703) was used to perform enzyme-linked immunoassays to measure sMUC5AC levels. Logistic regression, Cox regression models (univariate and multivariate), recurrence prediction, analysis of variance (ANOVA), t-tests, and Wilcoxon tests were used for statistical analysis. Results In the NAT cohort (n = 23), elevated sMUC5AC levels were significantly (P < 0.05) associated with pathological treatment response, margin positivity, and residual disease. Among 21 patients who had an R0/R1 resection (R2 resection, n=2), higher sMUC5AC levels were associated with shorter progression-free survival (PFS) (HR: 1.64, P = 0.0006) and overall survival (OS) (HR: 1.6, P = 0.005) on univariate analysis. Multivariate models confirmed sMUC5AC as an independent predictor of PFS and OS alongside pathological differentiation and postoperative therapy. Patients with lower sMUC5AC levels had more favorable pathological characteristics, better treatment responses, and improved survival outcomes. These findings were consistent in the FOLFIRINOX subgroup (n = 17). In the UpS cohort (n = 17), post-resection sMUC5AC levels tend to be associated with PFS (P = 0.07) and OS (P = 0.05). Combining sMUC5AC with Carbohydrate antigen (CA) 19-9 enhanced sensitivity (79%) and specificity (67%) to predict recurrence. Higher sMUC5AC levels were associated with earlier recurrence and poor survival outcomes, highlighting its utility in post-surgery risk stratification. Among patients with pre-treatment data (n = 11), sMUC5AC levels were significantly higher among patients with poorly differentiated tumors. Conclusion This study provides compelling evidence for the clinical utility of sMUC5AC as a prognostic biomarker among patients with resected PDA. Future large-scale studies are needed to validate these findings and establish standard thresholds for sMUC5AC integration into clinical practice.
Collapse
Affiliation(s)
- Ashish Manne
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center (OSUCCC), Columbus, OH, United States
| | - Yonghua Bao
- Clinical & Translational Science Shared Resource, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Ankur Sheel
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center (OSUCCC), Columbus, OH, United States
| | - Amir Sara
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center (OSUCCC), Columbus, OH, United States
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kannan Thanikachalam
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Ashwini Esnakula
- Department of Pathology, The Ohio State University Comprehensive Cancer Center (OSUCCC), Columbus, OH, United States
| | - Timothy M. Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center (OSUCCC), Columbus, OH, United States
| | - Jordan M. Cloyd
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center (OSUCCC), Columbus, OH, United States
| | - Susan Tsai
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center (OSUCCC), Columbus, OH, United States
| | - Anup Kasi
- Division of Medical Oncology, University of Kansas Cancer Center, Westwood, KS, United States
| | - Ravi Kumar Paluri
- Division of Hematology-Oncology, Department of Internal Medicine, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, United States
| | - Deepak Sherpally
- Department of Internal Medicine, New York Medical College, Valhalla, NY, United States
| | - Sravan Jeepalyam
- Department of Internal Medicine, Stormont Vail Health, Topeka, KS, United States
| | - Lianbo Yu
- Center of Biostatistics and Bioinformatics, The Ohio State University, Columbus, OH, United States
| | - Wancai Yang
- Clinical & Translational Science Shared Resource, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
45
|
Stoop TF, Javed AA, Oba A, Koerkamp BG, Seufferlein T, Wilmink JW, Besselink MG. Pancreatic cancer. Lancet 2025; 405:1182-1202. [PMID: 40187844 DOI: 10.1016/s0140-6736(25)00261-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 04/07/2025]
Abstract
Pancreatic cancer is frequently a lethal disease with an aggressive tumour biology often presenting with non-specific symptoms. Median survival is approximately 4 months with a 5-year survival of 13%. Surveillance is recommended in individuals with familial pancreatic cancer, specific mutations, and high-risk intraductal papillary mucinous neoplasm, as they are at high risk of developing pancreatic cancer. Chemotherapy combined with surgical resection remains the cornerstone of treatment. However, only a small subset of patients are candidates for surgery. Multi-agent chemotherapy has improved survival in the palliative setting for patients with metastatic disease, as (neo)adjuvant and induction therapy have in patients with borderline resectable and locally advanced pancreatic. Given that pancreatic cancer is predicted to become the second leading cause of cancer-related death by 2030, novel therapies are urgently needed.
Collapse
Affiliation(s)
- Thomas F Stoop
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Ammar A Javed
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands; Division of Surgical Oncology, Department of Surgery, New York University Medical Center, New York, NY, USA
| | - Atsushi Oba
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan; Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Thomas Seufferlein
- Department of International Medicine I, Ulm University Hospital, Ulm, Germany
| | - Johanna W Wilmink
- Department of Medical Oncology, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Marc G Besselink
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
46
|
Schwarz L, Bachet JB, Meurisse A, Bouché O, Assenat E, Piessen G, Hammel P, Regenet N, Taieb J, Turrini O, Paye F, Turpin A, Souche FR, Laurent C, Kianmanesh R, Michel P, Vernerey D, Mabrut JY, Turco C, Truant S, Sa Cunha A. Neoadjuvant FOLF(IRIN)OX Chemotherapy for Resectable Pancreatic Adenocarcinoma: A Multicenter Randomized Noncomparative Phase II Trial (PANACHE01 FRENCH08 PRODIGE48 study). J Clin Oncol 2025:JCO2401378. [PMID: 40184561 DOI: 10.1200/jco-24-01378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/10/2025] [Accepted: 02/12/2025] [Indexed: 04/06/2025] Open
Abstract
PURPOSE Despite limited RCTs, neoadjuvant chemotherapy (NAC) shows promise for resectable pancreatic adenocarcinoma (rPAC). Few prospective results are available on completing the full therapeutic sequence and oncologic outcomes with NAC. METHODS The PANACHE01-PRODIGE48 phase II trial randomly assigned 153 patients with rPAC (2:2:1) to four cycles of NAC (modified leucovorin, fluorouracil, irinotecan, and oxaliplatin [mFOLFIRINOX], arm 1; leucovorin, fluorouracil, and oxaliplatin [FOLFOX], arm 2) or up-front surgery (control) across 28 French centers (February 2017-July 2020). The primary objective was to evaluate the feasibility and efficacy of these NAC regimens. Two binary primary end points included 1-year overall survival (OS) postrandomization and the rate of patients completing the full therapeutic sequence. Event-free survival (EFS) assessed time to failure, defined as progression before surgery, unresectable/metastatic disease at surgery, recurrence, or death. RESULTS The primary objective was achieved for arm 1. In the intention-to-treat population, 70.8% (90% CI, 60.8 to 79.6) and 68% (90% CI, 55.5 to 78.8) completed the therapeutic sequence in arm 1 and arm 2, respectively. Within 12 months postrandomization, 84.3% (90% CI, 75.3 to 90.9) and 71.4% (90% CI, 59.0 to 81.8) of the patients were alive in arm 1 and arm 2, respectively. Treatment was safe and well-tolerated in both NAC arms. Arm 2 was stopped after interim analysis for lack of efficacy (H0 rejection for 1-year OS). One-year EFS rates were 51.4% (95% CI, 41.0 to 64.3), 43.1% (95% CI, 31.3 to 59.5), and 38.7% (95% CI, 24.1 to 62.0) in arm 1, arm 2, and control arm, respectively. CONCLUSION The feasibility and efficacy of mFOLFIRINOX in the perioperative setting are confirmed concerning therapeutic sequence completion and oncologic outcomes, supporting ongoing trials (PREOPANC3, Alliance AO21806). Further research is needed to identify patients who benefit from NAC (ClinicalTrials.gov identifier: NCT02959879; EudraCT: 2015-001851-65).
Collapse
Affiliation(s)
- Lilian Schwarz
- Department of Digestive Surgery, Rouen University Hospital, Rouen, France
- Department of Genomic and Personalized Medicine in Cancer and Neurological Disorders, UNIROUEN, UMR 1245 INSERM, Rouen University Hospital, Normandie University, Rouen, France
| | - Jean-Baptiste Bachet
- Department of Hepato-Gastroenterology, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Aurelia Meurisse
- Methodology and Quality of Life in Oncology Unit, Besançon University Hospital, Besançon, France
- EFS, INSERM, UMR RIGHT, Université de Franche-Comté, Besançon, France
| | - Olivier Bouché
- Digestive Oncology Department, Centre Hospitalier Universitaire Robert Debré, Reims, France
| | - Eric Assenat
- Medical Oncology Department, Centre Hospitalier Universitaire de Saint-Eloi, Montpellier, France
| | - Guillaume Piessen
- Department of Digestive and Oncological Surgery, Claude Huriez University Hospital, Chu Lille, France
| | - Pascal Hammel
- Digestive and Medical Oncology Department, Hôpital Paul Brousse and University Paris-Saclay, Villejuif, France
| | - Nicolas Regenet
- Department of Digestive Surgery, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Julien Taieb
- Department of Hepato-Gastroenterology, Georges Pompidou European Hospital, Carpem, Sorbonne Paris City, Paris Descartes University, Paris, France
| | - Olivier Turrini
- Department of Surgical Oncology, Institut Paoli-Calmettes, CRCM, Aix-Marseille University, Marseille, France
| | - Francois Paye
- Department of Surgery, Saint Antoine Hospital, Paris, France
- Bd de l'Hôpital, Sorbonne Université, Paris, France
| | - Anthony Turpin
- Department of Medical Oncology, Claude Huriez University Hospital, Chu Lille, France
| | - Francois-Regis Souche
- Department of Digestive Surgical Oncology, University Hospital of Montpellier, Montpellier, France
| | - Christophe Laurent
- Department of Hepato-Bilio-Pancreatic Surgery and Liver Transplantation, Haut Lévêque Hospital, CHU de Bordeaux, Pessac, France
| | - Reza Kianmanesh
- Digestive Surgery Department, Centre Hospitalier Universitaire Robert Debré, Reims, France
| | - Pierre Michel
- Department of Genomic and Personalized Medicine in Cancer and Neurological Disorders, UNIROUEN, UMR 1245 INSERM, Rouen University Hospital, Normandie University, Rouen, France
- Department of Digestive Oncology, Rouen University Hospital, Rouen, France
| | - Dewi Vernerey
- Methodology and Quality of Life in Oncology Unit, Besançon University Hospital, Besançon, France
- EFS, INSERM, UMR RIGHT, Université de Franche-Comté, Besançon, France
| | - Jean-Yves Mabrut
- Digestive Surgery and Liver Transplantation Department, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
- INSERM, CRCL UMR1052, Université de Lyon, Université Lyon 1, Lyon, France
| | - Celia Turco
- Digestive Surgery Department, University Hospital of Besançon, Besançon, France
| | - Stephanie Truant
- Department of Digestive Surgery and Liver Transplantation Department, CHRU Lille, CANTHER Laboratory Inserm UMR-S1277, University of Lille, Lille, France
| | - Antonio Sa Cunha
- Centre Hépato-Biliaire, Hôpital Universitaire Paul Brousse, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
47
|
Liaki V, Rosas-Perez B, Guerra C. Unlocking the Genetic Secrets of Pancreatic Cancer: KRAS Allelic Imbalances in Tumor Evolution. Cancers (Basel) 2025; 17:1226. [PMID: 40227826 PMCID: PMC11987834 DOI: 10.3390/cancers17071226] [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: 01/05/2025] [Revised: 03/13/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) belongs to the types of cancer with the highest lethality. It is also remarkably chemoresistant to the few available cytotoxic therapeutic options. PDAC is characterized by limited mutational heterogeneity of the known driver genes, KRAS, CDKN2A, TP53, and SMAD4, observed in both early-stage and advanced tumors. In this review, we summarize the two proposed models of genetic evolution of pancreatic cancer. The gradual or stepwise accumulated mutations model has been widely studied. On the contrary, less evidence exists on the more recent simultaneous model, according to which rapid tumor evolution is driven by the concurrent accumulation of genetic alterations. In both models, oncogenic KRAS mutations are the main initiating event. Here, we analyze the emerging topic of KRAS allelic imbalances and how it arises during tumor evolution, as it is often detected in advanced and metastatic PDAC. We also summarize recent evidence on how it affects tumor biology, metastasis, and response to therapy. To this extent, we highlight the necessity to include studies of KRAS allelic frequencies in the design of future therapeutic strategies against pancreatic cancer.
Collapse
Affiliation(s)
- Vasiliki Liaki
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
| | - Blanca Rosas-Perez
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
| | - Carmen Guerra
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
48
|
Zhou K, Liu Y, Tang C, Zhu H. Pancreatic Cancer: Pathogenesis and Clinical Studies. MedComm (Beijing) 2025; 6:e70162. [PMID: 40182139 PMCID: PMC11965705 DOI: 10.1002/mco2.70162] [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: 06/24/2024] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy, with pancreatic ductal adenocarcinoma (PDAC) being the most common and aggressive subtype, characterized by late diagnosis, aggressive progression, and resistance to conventional therapies. Despite advances in understanding its pathogenesis, including the identification of common genetic mutations (e.g., KRAS, TP53, CDKN2A, SMAD4) and dysregulated signaling pathways (e.g., KRAS-MAPK, PI3K-AKT, and TGF-β pathways), effective therapeutic strategies remain limited. Current treatment modalities including chemotherapy, targeted therapy, immunotherapy, radiotherapy, and emerging therapies such as antibody-drug conjugates (ADCs), chimeric antigen receptor T (CAR-T) cells, oncolytic viruses (OVs), cancer vaccines, and bispecific antibodies (BsAbs), face significant challenges. This review comprehensively summarizes these treatment approaches, emphasizing their mechanisms, limitations, and potential solutions, to overcome these bottlenecks. By integrating recent advancements and outlining critical challenges, this review aims to provide insights into future directions and guide the development of more effective treatment strategies for PC, with a specific focus on PDAC. Our work underscores the urgency of addressing the unmet needs in PDAC therapy and highlights promising areas for innovation in this field.
Collapse
Affiliation(s)
- Kexun Zhou
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Yingping Liu
- Department of RadiotherapyCancer HospitalChinese Academy of Medical SciencesBeijingChina
| | - Chuanyun Tang
- The First Clinical Medical College of Nanchang UniversityNanchang UniversityNanchangChina
| | - Hong Zhu
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengduChina
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
49
|
Takematsu T, Hayashi H, Ogawa D, Nakao Y, Yamao T, Kitano Y, Nakagawa S, Mima K, Baba Y, Baba H. Molecular Alterations Influencing the Prognostic Outcome in Small Pancreatic Cancer (≤2 cm). Pancreas 2025; 54:e295-e302. [PMID: 40262101 DOI: 10.1097/mpa.0000000000002430] [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: 06/09/2024] [Accepted: 11/01/2024] [Indexed: 04/24/2025]
Abstract
PURPOSE Pancreatic cancer (PC) is the most lethal cancer. The prognosis of small PC (tumor ≤ 20 mm) is better than that of larger PC, indicating the importance of detecting early-stage PC for patient outcome. The aim of this study was to elucidate the molecular features in small PC (≤20 mm). MATERIALS AND METHODS This study included 79 PC tumors (≤20 mm in pathological examination) resected between 2004 and 2022. c-Myc, Caveolin-1, Smad4, and Thrombospondin-1 were examined by immunostaining. These molecular alterations were compared in PC patients with tumor size ≤ 10 mm (n = 11) (14%) and 10 mm < tumor size ≤ 20 mm (n = 68) (86%). Mutation analyses of KRAS, PIK3CA, and BRAF were performed by pyrosequencing in 22 PCs. RESULTS PC with 10 mm < tumor size ≤ 20 mm showed significantly worse overall survival and disease-free survival than PC with tumor size < 10 mm (P = 0.024 and P = 0.028). Tumor c-Myc and stromal Caveolin-1 expressions were significantly increased in tumors larger than 10 mm (P = 0.02 and P = 0.04). c-Myc and Caveolin-1 expressions were associated with poor disease-free survival and overall survival. KRAS, PIK3CA, and BRAF mutation status did not differ between the 2 groups. CONCLUSIONS Tumor c-Myc and stromal Caveolin-1 overexpressions were detected in tumors larger than 10 mm. Their overexpressions were associated with worse prognosis even in small PC. These molecular alterations in small PC may be a clue for the detection of early-stage PC.
Collapse
Affiliation(s)
- Toru Takematsu
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
- Department of Gastroenterological Surgery, Imamura Hospital, Saga, Japan
| | - Hiromitsu Hayashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Daisuke Ogawa
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yosuke Nakao
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takanobu Yamao
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuki Kitano
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shigeki Nakagawa
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kosuke Mima
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
50
|
Wirth TC, Saborowski A, Kuehnle E, Fischer M, Bültmann E, von Kaisenberg C, Merten R. Chemo- and Radiotherapy of Gastrointestinal Tumors during Pregnancy. Visc Med 2025; 41:64-73. [PMID: 40201111 PMCID: PMC11975343 DOI: 10.1159/000540428] [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: 03/24/2024] [Accepted: 07/16/2024] [Indexed: 04/10/2025] Open
Abstract
Background Gastrointestinal cancers account for approximately one-fourth of all cancer cases and one-third of all tumor-related deaths worldwide. For the most frequent gastrointestinal tumor entities including colorectal, gastric, esophageal, and liver cancer, the incidence is expected to increase by more than 50% until 2040. While most gastrointestinal cancers are diagnosed beyond the age of fertility and predominantly in men, the increasing incidence of gastrointestinal malignancies in patients below the age of fifty suggests a growing importance in women of childbearing age. While localized cancers in pregnant women can either be monitored or treated surgically, more advanced stages might require radio- or chemotherapy to control tumor growth until delivery. Under these circumstances, critical decisions have to be made to preserve maternal health on the one side and minimize harm to the infant on the other side. Summary Here we summarize data from case reports, meta-analyses, and registries of women undergoing radio- or chemotherapy during pregnancy and provide guidance for therapeutic decision-making in pregnant women suffering from gastrointestinal cancers. Key Message After the first trimester, most chemotherapeutic regimens can be safely administered to pregnant patients with gastrointestinal cancers. With appropriate safety measures, both radiotherapy and radiochemotherapy can be applied to pregnant patients with rectal cancers.
Collapse
Affiliation(s)
- Thomas Christian Wirth
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Anna Saborowski
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Elna Kuehnle
- Department of Obstetrics and Gynaecology, Hannover Medical School, Hannover, Germany
| | - Mirko Fischer
- Department of Radiation Oncology, Hannover Medical School, Hannover, Germany
| | - Eva Bültmann
- Institute of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| | | | - Roland Merten
- Department of Radiation Oncology, Hannover Medical School, Hannover, Germany
| |
Collapse
|