101
|
Lansbergen MF, Dings MPG, Koster J, Labots M, Kerver ED, Jochems A, Homs MYV, de Vos-Geelen J, Hendriks MP, Tanck MWT, Wilmink JW, van Laarhoven HWM, Bijlsma MF. KRAS mutation status integrated with RNA subtyping improves prognostic modeling in FOLFIRINOX-treated metastatic pancreatic cancer. MED 2025:100601. [PMID: 39938521 DOI: 10.1016/j.medj.2025.100601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/05/2024] [Accepted: 01/14/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND First-line chemotherapy (5-fluorouracil, leucovorin, irinotecan, and oxaliplatin [FOLFIRINOX]) benefits few patients with metastatic pancreatic ductal adenocarcinoma (mPDAC). Prognostic markers for treatment-related survival are needed. This study validated the added benefit of whole-genome sequencing (WGS) to transcriptome-based classification in modeling FOLFIRINOX-related survival. METHODS Patients with mPDAC planning to start FOLFIRINOX were included in a prospective nationwide cohort. Pretreatment biopsies were submitted to WGS and RNA sequencing. Samples of non-FOLFIRINOX-treated patients were included for exploratory analyses. FINDINGS WGS was performed in biopsies from 108 FOLFIRINOX-treated patients and 51 non-FOLFIRINOX-treated patients. 12% of the tumors were KRAS wild type. These tumors had more targetable alterations (42% vs. 17%) and were associated with a longer median overall survival (mOS) than KRAS mutant tumors (7.8 months in KRAS mutant vs. 17.7 months in wild-type tumors, p = 0.0024). Transcriptome-based clustering revealed a tumor subgroup showing low classical and basal-like gene expression, enriched for KRAS wild-type status (p < 0.0001), a so-called "classifier-negative" subtype. The gene expression of these classifier-negative tumors correlated with neural-like signatures. For patients with a homologous recombination-deficient (HRD) tumor, mOS was not increased (8.0 months in homologous recombination-proficient [HRP] vs. 13.3 months in HRD tumors, p = 0.21). CONCLUSIONS KRAS wild-type tumors are a distinct PDAC subgroup with a better prognosis. Consequently, KRAS status assessment before transcriptome-based subtyping can stratify patients into three prognostic molecular subgroups (KRAS wild type, KRAS mutant classical, and KRAS mutant basal like). This integrative way of classification should be validated prior to incorporation in diagnostic practice. FUNDING ZonMw "Good Use of Medicine" program (848101012).
Collapse
Affiliation(s)
- Marjolein F Lansbergen
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, 1081 HV Amsterdam, the Netherlands; Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, University of Amsterdam, 1081 BT Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, 1081 BT Amsterdam, the Netherlands
| | - Mark P G Dings
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, University of Amsterdam, 1081 BT Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, 1081 BT Amsterdam, the Netherlands
| | - Jan Koster
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, University of Amsterdam, 1081 BT Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, 1081 BT Amsterdam, the Netherlands
| | - Mariette Labots
- Cancer Center Amsterdam, Imaging and Biomarkers, 1081 BT Amsterdam, the Netherlands; Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, the Netherlands
| | - Emile D Kerver
- Department of Medical Oncology, OLVG, 1091 AC Amsterdam, the Netherlands
| | - Anouk Jochems
- Department of Internal Medicine, Haaglanden Medical Center, 2512 VA The Hague, the Netherlands
| | - Marjolein Y V Homs
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, the Netherlands
| | - Judith de Vos-Geelen
- Department of Medical Oncology, GROW - Research Institute for Oncology & Reproduction, Maastricht University Medical Center, 6229 HX Maastricht, the Netherlands
| | - Mathijs P Hendriks
- Department of Medical Oncology, Northwest Clinics, 1815 JD Alkmaar, the Netherlands
| | - Michael W T Tanck
- Department of Epidemiology and Data Science, Amsterdam Public Health, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Johanna W Wilmink
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, 1081 HV Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, 1081 BT Amsterdam, the Netherlands
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, 1081 HV Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, 1081 BT Amsterdam, the Netherlands
| | - Maarten F Bijlsma
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, University of Amsterdam, 1081 BT Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, 1081 BT Amsterdam, the Netherlands.
| |
Collapse
|
102
|
Zhang N, Chen S, Shi Y, Wang Z, Jia R, Dai G. Charlson syndrome index predicted survival in pancreatic cancer patients received immunotherapy. Front Immunol 2025; 16:1487318. [PMID: 39963141 PMCID: PMC11831428 DOI: 10.3389/fimmu.2025.1487318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 01/03/2025] [Indexed: 02/20/2025] Open
Abstract
Objective The Charlson Comorbidity Index (CCI) is a widely utilized assessment tool for evaluating the mortality rate among patients with chronic diseases and tumors. Currently, there is a dearth of research investigating the correlation between CCI and survival rates in advanced pancreatic cancer patients received immunotherapy. Therefore, this study aims to elucidate the association between CCI and survival rates in real-world settings for pancreatic cancer patients received immunotherapy. Methods A total of 104 patients with advanced pancreatic cancer who received immunotherapy at the General Hospital of the People's Liberation Army between September 2015 and September 2020 were included in this study. The patients were categorized into two groups based on their Charlson Comorbidity Index (CCI) scores: low CCI group (CCI <7) and high CCI group (CCI ≥7). The statistical analysis focused on examining the correlation between CCI score and survival outcome. Results The high CCI group exhibited significantly lower overall survival (OS) and progression-free survival (PFS) compared to the low CCI group (p<0.05). The median OS for the high CCI and low CCI groups were 7.82 and 44.17 months, respectively, while the median PFS were 2.40 and 6.40 months, respectively. Multivariate analysis revealed that high CCI was independently risk factor for both OS (HR=2.801, 95%CI: 1.433-5.472, p=0.003) and PFS (HR=2.546, 95%CI: 1.389-4.668, p=0.003). Conclusion The CCI score serves as a significant independent predictive indicator for advanced pancreatic cancer patients received immunotherapy.
Collapse
Affiliation(s)
- Nan Zhang
- Medical School of Chinese PLA, Beijing, China
- Department of Medical Oncology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shiyun Chen
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yue Shi
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhikuan Wang
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ru Jia
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Guanghai Dai
- Department of Medical Oncology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
103
|
Ma Y, Dumesny C, Dong L, Ang CS, Nikfarjam M, He H. Knockout of p21-Activated Kinase 4 Stimulates MHC I Expression of Pancreatic Cancer Cells via an Autophagy-Independent Pathway. Cancers (Basel) 2025; 17:511. [PMID: 39941877 PMCID: PMC11817421 DOI: 10.3390/cancers17030511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pancreatic ductal adenocarcinoma (PDA) is one of the most malignant solid cancers. KRAS mutation accounts for over 90% of cases. p21-activated kinases (PAKs) act downstream of KRAS and are involved in tumorigenesis. The inhibition of PAK4 suppresses PDA by stimulating the tumor infiltration of cytotoxic T cells. The major histocompatibility complex class I (MHC I) is a key in presenting antigens to cytotoxic T cells. MHC I degradation via autophagy promotes the immune evasion of pancreatic cancer. We investigated the effect of PAK4 inhibition on MHC I expression and autophagy. METHODS In this study, using proteomic analysis, fluorescence-activated cell sorting (FACS), and immunoblotting, we examined the effect of PAK4 knockout (KO) in human PDA cells on the expression of MHC I and autophagy to identify the mechanism involved in the stimulation of cytotoxic T cells by PAK4 inhibition. RESULTS We found that PAK4 KO increased MHC I expression in two human PDA cell lines: MiaPaCa-2 and PANC-1. PAK4 KO also increased cancer cell autophagy. However, the inhibition of autophagy by chloroquine (CQ) did not affect the effect of PAK4 KO on apoptosis and cell death. More importantly, the inhibition of autophagy by CQ did not alter the expression of MHC I stimulated by PAK4 KO, indicating that PAK4 KO stimulated MHC I expression via an autophagy-independent pathway. CONCLUSIONS We identified a role of PAK4 in MHC I expression by PDA cells, which is independent of autophagy.
Collapse
Affiliation(s)
- Yi Ma
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
- Department of General Surgery, Monash Health, Clayton, VIC 3806, Australia
| | - Chelsea Dumesny
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
| | - Li Dong
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
| | - Ching-Seng Ang
- Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3052, Australia;
| | - Mehrdad Nikfarjam
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
- Department of Hepato-Pancreato-Biliary Surgery, Austin Health, Heidelberg, VIC 3084, Australia
| | - Hong He
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
| |
Collapse
|
104
|
Melle F, Menon D, Conniot J, Ostolaza-Paraiso J, Mercado S, Oliveira J, Chen X, Mendes BB, Conde J, Fairen-Jimenez D. Rational Design of Metal-Organic Frameworks for Pancreatic Cancer Therapy: from Machine Learning Screening to In Vivo Efficacy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2412757. [PMID: 39895194 DOI: 10.1002/adma.202412757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/09/2024] [Indexed: 02/04/2025]
Abstract
Despite improvements in cancer survival rates, metastatic and surgery-resistant cancers, such as pancreatic cancer, remain challenging, with poor prognoses and limited treatment options. Enhancing drug bioavailability in tumors, while minimizing off-target effects, is crucial. Metal-organic frameworks (MOFs) have emerged as promising drug delivery vehicles owing to their high loading capacity, biocompatibility, and functional tunability. However, the vast chemical diversity of MOFs complicates the rational design of biocompatible materials. This study employed machine learning and molecular simulations to identify MOFs suitable for encapsulating gemcitabine, paclitaxel, and SN-38, and identified PCN-222 as an optimal candidate. Following drug loading, MOF formulations are improved for colloidal stability and biocompatibility. In vitro studies on pancreatic cancer cell lines have shown high biocompatibility, cellular internalization, and delayed drug release. Long-term stability tests demonstrated a consistent performance over 12 months. In vivo studies in pancreatic tumor-bearing mice revealed that paclitaxel-loaded PCN-222, particularly with a hydrogel for local administration, significantly reduced metastatic spread and tumor growth compared to the free drug. These findings underscore the potential of PCN-222 as an effective drug delivery system for the treatment of hard-to-treat cancers.
Collapse
Affiliation(s)
- Francesca Melle
- The Adsorption & Advanced Materials Laboratory (AAML), Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Dhruv Menon
- The Adsorption & Advanced Materials Laboratory (AAML), Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - João Conniot
- NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC), NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Jon Ostolaza-Paraiso
- The Adsorption & Advanced Materials Laboratory (AAML), Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Sergio Mercado
- The Adsorption & Advanced Materials Laboratory (AAML), Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Jhenifer Oliveira
- NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC), NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Xu Chen
- The Adsorption & Advanced Materials Laboratory (AAML), Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Bárbara B Mendes
- NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC), NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - João Conde
- NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC), NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - David Fairen-Jimenez
- The Adsorption & Advanced Materials Laboratory (AAML), Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| |
Collapse
|
105
|
Pinto F, Roldão M, Ribeiro AM, Oliveira J, Bento I. From Persisting Diabetes to the Diagnosis of Pancreatic Cancer: A Case Report. Cureus 2025; 17:e79408. [PMID: 40125183 PMCID: PMC11930273 DOI: 10.7759/cureus.79408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2025] [Indexed: 03/25/2025] Open
Abstract
The diagnosis of new-onset diabetes mellitus (DM) in individuals with a consumptive syndrome warrants a comprehensive and systematic etiological investigation. This article presents the case of a 45-year-old male, smoker, with a history of a treated C hepatitis virus, diagnosed with type 2 diabetes, accompanied by complaints of fatigue, anorexia, and unintentional weight loss. Despite an initial workup by the general practitioner, including colonoscopy, endoscopy, and abdominal-pelvic CT scan, no neoplastic etiology was identified. A few months later, due to worsening pain and continued weight loss, along with multiple visits to the emergency room, he was referred for an internal medicine consultation for further evaluation. During this investigation, stage IV pancreatic cancer was diagnosed, with multiple organ involvement, and the patient was indicated for best supportive care. The patient passed away four months after the diagnosis. Through this clinical case, the authors aim to highlight that in cases of newly diagnosed diabetes mellitus with poor control despite therapy and negative imaging results, and associated risk factors, such as tobacco and virus C hepatitis, continuous etiological investigation is crucial, particularly when there is a failure to achieve metabolic control despite appropriate pharmacological treatment and diet.
Collapse
Affiliation(s)
- Fausto Pinto
- Internal Medicine, Hospital São Francisco Xavier, Lisbon, PRT
| | - Marta Roldão
- Internal Medicine, Hospital São Francisco Xavier, Lisbon, PRT
| | | | - João Oliveira
- Internal Medicine, Hospital São Francisco Xavier, Lisbon, PRT
| | - Ines Bento
- Internal Medicine, Hospital São Francisco Xavier, Lisbon, PRT
| |
Collapse
|
106
|
Sato R, Hotta K, Kubo T, Horiguchi S, Kato H, Matsumoto K, Kozuki T, Udono H, Kiura K, Otsuka M. Durable Response to Nivolumab Combined With Metformin in Advanced Pancreatic Cancer: A Case Report With Seven Years of Follow-Up. Cureus 2025; 17:e79001. [PMID: 40092004 PMCID: PMC11910964 DOI: 10.7759/cureus.79001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
We report a case of poorly differentiated pancreatic cancer that showed an exceptional response to combination therapy with nivolumab and metformin. A 58-year-old man presented with epigastric pain and was diagnosed with locally advanced pancreatic cancer with para-aortic lymph node metastasis. After disease progression following modified FOLFIRINOX therapy (a combination of fluorouracil, leucovorin, irinotecan, and oxaliplatin), the patient was enrolled in a phase Ib clinical trial of nivolumab (3 mg/kg biweekly) combined with metformin (750 mg/day). Post-treatment imaging showed marked tumor shrinkage with normalization of the tumor markers. During treatment, the patient was diagnosed with early-stage lung cancer and underwent successful left S1+S2 segmentectomy with temporary suspension of immunotherapy. The therapeutic response of pancreatic cancer has been sustained for seven years, with minimal residual disease. This unprecedented response duration is particularly noteworthy considering his microsatellite stability, which typically predicts a limited response to immune checkpoint inhibition. This case demonstrates an exceptional response to nivolumab and metformin combination therapy in poorly differentiated pancreatic cancer. The remarkable durability of the response suggests the need for further investigation to identify patients most likely to benefit from this therapeutic approach.
Collapse
Affiliation(s)
- Ryosuke Sato
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama, JPN
| | - Katsuyuki Hotta
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, JPN
| | - Toshio Kubo
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, JPN
| | - Shigeru Horiguchi
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama, JPN
| | - Hironari Kato
- Department of Gastroenterology, Okayama City Hospital, Okayama, JPN
| | - Kazuyuki Matsumoto
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama, JPN
| | - Toshiyuki Kozuki
- Department of Respiratory Medicine and Allergology, Kochi Medical School, Kochi University, Kochi, JPN
| | - Heiichiro Udono
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, JPN
| | - Katsuyuki Kiura
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, JPN
| | - Motoyuki Otsuka
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama, JPN
| |
Collapse
|
107
|
Kurdyn A, Pawłowska M, Paluszkiewicz E, Cichorek M, Augustin E. c-Myc inhibition and p21 modulation contribute to unsymmetrical bisacridines-induced apoptosis and senescence in pancreatic cancer cells. Pharmacol Rep 2025; 77:182-209. [PMID: 39361216 PMCID: PMC11743403 DOI: 10.1007/s43440-024-00658-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 01/21/2025]
Abstract
BACKGROUND Pancreatic cancer (PC) is one of the most aggressive cancers and is the seventh leading cause of cancer-related death worldwide. PC is characterized by rapid progression and resistance to conventional treatments. Mutations in KRAS, CDKN2A, TP53, SMAD4/DPC4, and MYC are major genetic alterations associated with poor treatment outcomes in patients with PC. Therefore, optimizing PC therapy is a tremendous challenge. Unsymmetrical bisacridines (UAs), synthesized by our group, are new promising compounds that have exhibited high cytotoxicity and antitumor activity against several solid tumors, including pancreatic cancer. METHODS The cellular effects induced by UAs in PC cells were evaluated by MTT assay (cell growth inhibition), flow cytometry, and fluorescence and light microscopy (cell cycle distribution, apoptosis, and senescence detection). Analysis of the effects of UAs on the levels of proteins (c-Myc, p53, SMAD4, p21, and p16) was performed by Western blotting. RESULTS Apoptosis was the main triggered mechanism of death after UAs treatment, and induction of the SMAD4 protein can facilitate this process. c-Myc, which is one of the molecular targets of UAs, can participate in the induction of cell death in a p53-independent manner. Moreover, UAs can also induce accelerated senescence through the upregulation of p21. Notably, senescent cells can die via apoptosis after prolonged exposure to UAs. CONCLUSIONS UAs have emerged as potent anticancer agents that induce apoptosis by inhibiting c-Myc protein and triggering cellular senescence in a dose-dependent manner by increasing p21 levels. Thus, UAs exhibit desirable features as promising candidates for future pancreatic anticancer therapies.
Collapse
Affiliation(s)
- Agnieszka Kurdyn
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, Gdańsk, 80-233, Poland
| | - Monika Pawłowska
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, Gdańsk, 80-233, Poland
| | - Ewa Paluszkiewicz
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, Gdańsk, 80-233, Poland
| | - Mirosława Cichorek
- Department of Embryology, Medical University of Gdańsk, Dębinki 1, Gdańsk, 80-211, Poland
| | - Ewa Augustin
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, Gdańsk, 80-233, Poland.
| |
Collapse
|
108
|
Aberle MR, Coolsen MME, Wenmaekers G, Volmer L, Brecheisen R, van Dijk D, Wee L, Van Dam RM, de Vos-Geelen J, Rensen SS, Damink SWMO. Skeletal muscle is independently associated with grade 3-4 toxicity in advanced stage pancreatic ductal adenocarcinoma patients receiving chemotherapy. Clin Nutr ESPEN 2025; 65:134-143. [PMID: 39577693 DOI: 10.1016/j.clnesp.2024.11.004] [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: 07/01/2024] [Revised: 11/06/2024] [Accepted: 11/09/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Patients with advanced-stage pancreatic ductal adenocarcinoma (PDAC) are regularly treated with FOLFIRINOX, a chemotherapy regimen based on 5-fluorouracil, irinotecan and oxaliplatin, which is associated with high toxicity. Dosing of FOLFIRINOX is based on body surface area, risking under- or overdosing caused by altered pharmacokinetics due to interindividual differences in body composition. This study aimed to investigate the relationship between body composition and treatment toxicity in advanced stage PDAC patients treated with FOLFIRINOX. METHODS Data from patients treated at the Maastricht University Medical Centre + between 2012 and 2020 were collected retrospectively (n = 65). Skeletal muscle-, visceral adipose tissue, subcutaneous adipose tissue-, (SM-Index, VAT-Index, SAT-Index resp.) and Skeletal Muscle Radiation Attenuation (SM-RA) were calculated after segmentation of computed tomography (CT) images at the third lumbar level using a validated deep learning method. Lean body mass (LBM) was estimated using SM-Index. Toxicities were scored and grade 3-4 adverse events were considered dose-limiting toxicities (DLTs). RESULTS Sixty-seven DLTs were reported during the median follow-up of 51.4 (95%CI 39.2-63.7) weeks. Patients who experienced at least one DLT had significantly higher dose intensity per LBM for all separate cytotoxics of FOLFIRINOX. Independent prognostic factors for the number of DLTs per cycle were: sarcopenia (β = 0.292; 95%CI 0.013 to 0.065; p = 0.013), SM-Index change (% per 30 days, β = -0.045; 95%CI -0.079 to -0.011; p = 0.011), VAT-Index change (% per 30 days, β = -0.006; 95%CI -0.012 to 0.000; p = 0.040) between diagnosis and the first follow-up CT scan, and cumulative relative dose intensity >80 % (β = -0.315; 95 % CI -0.543 to -0.087; p = 0.008). CONCLUSION Sarcopenia and early muscle and fat wasting during FOLFIRINOX treatment were associated with treatment-related toxicity, warranting exploration of body composition guided personalized dosing of chemotherapeutics to limit DLTs.
Collapse
Affiliation(s)
- Merel R Aberle
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands; Department of Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Mariëlle M E Coolsen
- Department of Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of General, Visceral- and Transplantation Surgery, RWTH Aachen University, Germany
| | - Gilles Wenmaekers
- Department of Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Leroy Volmer
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands; Department of Radiotherapy (MAASTRO), Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Ralph Brecheisen
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands; Department of Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - David van Dijk
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands; Department of Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Leonard Wee
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands; Department of Radiotherapy (MAASTRO), Maastricht University Medical Centre+, Maastricht, the Netherlands; Clinical Data Science, Maastricht University, Maastricht, the Netherlands
| | - Ronald M Van Dam
- Department of Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands; Department of General, Visceral- and Transplantation Surgery, RWTH Aachen University, Germany
| | - Judith de Vos-Geelen
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands; Department of Medical Oncology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Sander S Rensen
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands.
| | - Steven W M Olde Damink
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands; Department of Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of General, Visceral- and Transplantation Surgery, RWTH Aachen University, Germany
| |
Collapse
|
109
|
Beutel AK, Ekizce M, Ettrich TJ, Seufferlein T, Lindenmayer J, Gout J, Kleger A. Organoid-based precision medicine in pancreatic cancer. United European Gastroenterol J 2025; 13:21-33. [PMID: 39540683 PMCID: PMC11866314 DOI: 10.1002/ueg2.12701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/05/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) ranks among the leading causes of cancer-related deaths worldwide. Despite advances in precision oncology in other malignancies, treatment of PDAC still largely relies on conventional chemotherapy. Given the dismal prognosis and heterogeneity in PDAC, there is an urgent need for personalized therapeutic strategies to improve treatment response. Organoids, generated from patients' tumor tissue, have emerged as a powerful tool in cancer research. These three-dimensional models faithfully recapitulate the morphological and genetic features of the parental tumor and retain patient-specific heterogeneity. This review summarizes existing precision oncology approaches in PDAC, explores current applications and limitations of organoid cultures in personalized medicine, details preclinical studies correlating in vitro organoid prediction and patient treatment response, and provides an overview of ongoing organoid-based clinical trials.
Collapse
Affiliation(s)
- Alica K. Beutel
- Department of Internal Medicine IUniversity Hospital UlmUlmGermany
| | - Menar Ekizce
- Institute of Molecular Oncology and Stem Cell BiologyUlm University HospitalUlmGermany
| | | | | | | | - Johann Gout
- Institute of Molecular Oncology and Stem Cell BiologyUlm University HospitalUlmGermany
| | - Alexander Kleger
- Institute of Molecular Oncology and Stem Cell BiologyUlm University HospitalUlmGermany
- Core Facility OrganoidsMedical Faculty of Ulm UniversityUlmGermany
- Division of Interdisciplinary PancreatologyDepartment of Internal Medicine IUlm University HospitalUlmGermany
| |
Collapse
|
110
|
Varghese AM, Perry MA, Chou JF, Nandakumar S, Muldoon D, Erakky A, Zucker A, Fong C, Mehine M, Nguyen B, Basturk O, Balogun F, Kelsen DP, Brannon AR, Mandelker D, Vakiani E, Park W, Yu KH, Stadler ZK, Schattner MA, Jarnagin WR, Wei AC, Chakravarty D, Capanu M, Schultz N, Berger MF, Iacobuzio-Donahue CA, Bandlamudi C, O'Reilly EM. Clinicogenomic landscape of pancreatic adenocarcinoma identifies KRAS mutant dosage as prognostic of overall survival. Nat Med 2025; 31:466-477. [PMID: 39753968 PMCID: PMC11835752 DOI: 10.1038/s41591-024-03362-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/17/2024] [Indexed: 01/11/2025]
Abstract
Nearly all pancreatic adenocarcinomas (PDAC) are genomically characterized by KRAS exon 2 mutations. Most patients with PDAC present with advanced disease and are treated with cytotoxic therapy. Genomic biomarkers prognostic of disease outcomes have been challenging to identify. Herein leveraging a cohort of 2,336 patients spanning all disease stages, we characterize the genomic and clinical correlates of outcomes in PDAC. We show that a genomic subtype of KRAS wild-type tumors is associated with early disease onset, distinct somatic and germline features, and significantly better overall survival. Allelic imbalances at the KRAS locus are widespread. KRAS mutant allele dosage gains, observed in one in five (20%) KRAS-mutated diploid tumors, are correlated with advanced disease and demonstrate prognostic potential across disease stages. With the rapidly expanding landscape of KRAS targeting, our findings have potential implications for clinical practice and for understanding de novo and acquired resistance to RAS therapeutics.
Collapse
Affiliation(s)
- Anna M Varghese
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Maria A Perry
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Joanne F Chou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Subhiksha Nandakumar
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Daniel Muldoon
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Amanda Erakky
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Amanda Zucker
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Christopher Fong
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Miika Mehine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Bastien Nguyen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Olca Basturk
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Fiyinfolu Balogun
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - David P Kelsen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - A Rose Brannon
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Diana Mandelker
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Efsevia Vakiani
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Wungki Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Kenneth H Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Mark A Schattner
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - William R Jarnagin
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Alice C Wei
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Debyani Chakravarty
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Marinela Capanu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Nikolaus Schultz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Michael F Berger
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Christine A Iacobuzio-Donahue
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Chaitanya Bandlamudi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| |
Collapse
|
111
|
Ko SW, Jo IH, Yoon SB. Feasibility and clinical utility of endoscopic ultrasound-guided tissue acquisition for comprehensive genomic profiling in pancreatic cancer: A systematic review and meta-analysis. Pancreatology 2025; 25:89-97. [PMID: 39732591 DOI: 10.1016/j.pan.2024.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/09/2024] [Accepted: 12/21/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND Endoscopic ultrasound-guided tissue acquisition (EUS-TA) has become essential for diagnosing pancreatic ductal adenocarcinoma (PDAC) and is increasingly utilized for comprehensive genome profiling (CGP) to advance precision medicine. This systematic review and meta-analysis assess the feasibility and clinical utility of EUS-TA samples for CGP in PDAC. METHODS We conducted a thorough systematic literature search in PubMed, EMBASE, and the Cochrane Library up to October 2023. Key outcomes included sequencing success rates, detection rates of four major driver genes and actionable genes, and concordance rates with other sample types or methodologies. RESULTS A total of 23 studies met the inclusion criteria. The pooled sequencing success rate was 83.9 % [95 % confidence interval (CI): 75.8-89.7 %]. No significant difference was observed in sequencing success rates between fine needle aspiration and biopsy (odds ratio 1.77, 95 % CI 0.70-4.47). Meta-regression analysis revealed that the minimum DNA requirement for CGP significantly influenced sequencing success rates. The pooled mutation rate for K-ras was 86.4 % (95 % CI 83.6-88.8), while potentially actionable mutations had a pooled rate of 17.7 % (95 % CI 12.8-23.8). The concordance rate between CGP results from EUS-guided samples and surgical specimens was 81.6 % (95 % CI 68.2-90.1). CONCLUSION Comprehensive genomic profiling of PDAC using EUS-TA-derived samples demonstrated feasibility in clinical settings. Approximately 18 % of patients undergoing CGP exhibited potentially actionable mutations, highlighting the potential for personalized therapeutic approaches.
Collapse
Affiliation(s)
- Sung Woo Ko
- Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ik Hyun Jo
- Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, South Korea
| | - Seung Bae Yoon
- Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea.
| |
Collapse
|
112
|
Madani SP, Mirza-Aghazadeh-Attari M, Mohseni A, Afyouni S, Zandieh G, Shahbazian H, Borhani A, Yazdani Nia I, Laheru D, Pawlik TM, Kamel IR. Value of radiomics features extracted from baseline computed tomography images in predicting overall survival in patients with nonsurgical pancreatic ductal adenocarcinoma: incorporation of a radiomics score to a multiparametric nomogram to predict 1-year overall survival. J Gastrointest Surg 2025; 29:101882. [PMID: 39528002 DOI: 10.1016/j.gassur.2024.101882] [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: 06/29/2024] [Revised: 10/01/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE This study aimed to determine the value of radiomics features derived from baseline computed tomography (CT) scans and volumetric measurements to predict overall survival (OS) in patients with nonsurgical pancreatic ductal adenocarcinoma (PDAC) treated with a chemotherapy combination regimen of 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX). METHODS In this retrospective single-institution study, 131 patients with nonsurgical PDAC who received FOLFIRINOX neoadjuvant chemotherapy between December 2012 and November 2021 were included. Pretreatment contrast-enhanced CT images were obtained for all patients before inclusion. The primary tumor was contoured by an expert radiologist with 25 years of experience. A total of 845 radiomics features, including first-, second-, and higher-order features, were extracted from the total tumor volume. A feature reduction pipeline was used to reduce the dimensionality of the data. The selected features were used to generate a radiomics score based on the Least Absolute Shrinkage and Selection Operator coefficients. A high-dimensional Cox model was generated on the basis of the radiomics score and other quantitative and semantic imaging findings. RESULTS From the 845 radiomics features extracted, 45 were significantly different between the tertiles. The following equation was used to generate a radiomics score: radiomics score = SmallAreaEmphasis (-66.87801 + LargeDependenceEmphasis) - 0.2345916. The radiomics score was significantly different among the 3 groups of the radiomics features (P = .034). The overall difference in survival was significant among the 3 groups (P = .02). The nomogram showed good calibration and showed significant differences among the patients when they were classified as tertiles (P < .00). CONCLUSION Radiomics approaches have the potential to predict OS in nonsurgical patients with PDAC, and the inclusion of semantic imaging findings and pathologic data could further enhance prognostication in patients with PDAC.
Collapse
Affiliation(s)
- Seyedeh Panid Madani
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, United States
| | | | - Alireza Mohseni
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, United States
| | - Shadi Afyouni
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, United States
| | - Ghazal Zandieh
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, United States
| | - Haneyeh Shahbazian
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, United States
| | - Ali Borhani
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, United States
| | - Iman Yazdani Nia
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, United States
| | - Daniel Laheru
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Timothy M Pawlik
- Department of Surgery, Wexner Medical Center, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Ihab R Kamel
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, United States; Department of Radiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
113
|
Xu J, Pham MD, Corbo V, Ponz-Sarvise M, Oni T, Öhlund D, Hwang CI. Advancing pancreatic cancer research and therapeutics: the transformative role of organoid technology. Exp Mol Med 2025; 57:50-58. [PMID: 39814914 PMCID: PMC11799150 DOI: 10.1038/s12276-024-01378-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 01/18/2025] Open
Abstract
Research on pancreatic cancer has transformed with the advent of organoid technology, providing a better platform that closely mimics cancer biology in vivo. This review highlights the critical advancements facilitated by pancreatic organoid models in understanding disease progression, evaluating therapeutic responses, and identifying biomarkers. These three-dimensional cultures enable the proper recapitulation of the cellular architecture and genetic makeup of the original tumors, providing insights into the complex molecular and cellular dynamics at various stages of pancreatic ductal adenocarcinoma (PDAC). We explore the applications of pancreatic organoids in dissecting the tumor microenvironment (TME); elucidating cancer progression, metastasis, and drug resistance mechanisms; and personalizing therapeutic strategies. By overcoming the limitations of traditional 2D cultures and animal models, the use of pancreatic organoids has significantly accelerated translational research, which is promising for improving diagnostic and therapeutic approaches in clinical settings, ultimately aiming to improve the outcomes of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Jihao Xu
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Minh Duc Pham
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Mariano Ponz-Sarvise
- Department of Medical Oncology and Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Pamplona, Spain
| | - Tobiloba Oni
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Daniel Öhlund
- Umeå University, Department of Diagnostics and Intervention, and Wallenberg Centre for Molecular Medicine at Umeå University, Umeå, Sweden
| | - Chang-Il Hwang
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA.
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, 95817, USA.
| |
Collapse
|
114
|
Tanisaka Y, Ryozawa S, Mizuide M, Fujita A, Jinushi R, Watanabe R, Sato R, Yasuda M. The correlation between tumoral CD8 expression and clinical course in patients with unresectable pancreatic cancer using tissue samples acquired by endoscopic ultrasound-guided tissue acquisition. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2025; 32:132-138. [PMID: 39328021 DOI: 10.1002/jhbp.12072] [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: 09/28/2024]
Abstract
BACKGROUND The aim of this study was to evaluate the correlation between tumoral CD8 expression and the clinical course in patients with unresectable pancreatic ductal adenocarcinoma (PDAC) using tissue samples acquired by endoscopic ultrasound-guided tissue acquisition (EUS-TA). METHODS Patients with unresectable PDAC who underwent EUS-TA prior to treatment between September 2017 and October 2021 were included. The localization of the CD8-positive areas was qualitatively evaluated. We divided the patients into high and low groups based on the median percentage of CD8-positive areas. The correlation between the number of CD8-positive areas and overall survival was assessed. Furthermore, the response to chemotherapy was assessed in patients who underwent chemotherapy. RESULTS A total of 169 patients were included in the analysis. The median overall survival was 171 days (95% confidence interval [CI]: 86-401). The median CD8-positive area was 0.10% (95% CI: 0.05-0.26). The median overall survival in the high (≥0.1%) and low (<0.1%) CD8-positive groups were 156 and 213.5 days, respectively (p = .33). The number of CD8-positive areas was not correlated with the overall survival and response to chemotherapy (p = .69). CONCLUSIONS Tissue samples acquired using EUS-TA from patients with unresectable PDAC showed that CD8 expression did not affect the clinical course of patients.
Collapse
Affiliation(s)
- Yuki Tanisaka
- Department of Gastroenterology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Shomei Ryozawa
- Department of Gastroenterology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Masafumi Mizuide
- Department of Gastroenterology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Akashi Fujita
- Department of Gastroenterology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Ryuhei Jinushi
- Department of Gastroenterology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Ryuichi Watanabe
- Department of Gastroenterology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Ryo Sato
- Department of Gastroenterology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Masanori Yasuda
- Department of Pathology, Saitama Medical University International Medical Center, Hidaka, Japan
| |
Collapse
|
115
|
Saito K, Nakai Y, Sasaki T, Takeda T, Ueno M, Tezuka S, Isayama H, Tomishima K, Kojima Y, Yamamoto N, Ito Y, Oyama H, Toda N, Takagi K, Matsubara S, Mohri D, Sato T, Fujishiro M. Impact of Statin Use on Survival in Patients With Unresectable Pancreatic cancer Receiving Gemcitabine Plus Nab-Paclitaxel: A Multicenter Retrospective Study. Pancreas 2025; 54:e107-e113. [PMID: 39928888 DOI: 10.1097/mpa.0000000000002411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
PURPOSE The aim of this multicenter retrospective study was to evaluate the impact of statin use on clinical outcomes in patients with unresectable pancreatic cancer (PC) receiving gemcitabine plus nab-paclitaxel (GnP) in a large Japanese cohort. MATERIALS AND METHODS We retrospectively reviewed the medical records including data on the use of concomitant medications in patients with unresectable PC receiving GnP between January 2015 and January 2019 at 10 hospitals. Prognostic factors for progression-free survival (PFS) and overall survival (OS) were evaluated. RESULTS A total of 1682 patients were included in the analysis; of which 322 patients (19%) received statins and 1360 (81%) did not receive statin. The median PFS and OS were 7.5 versus 7.3 months (P = 0.87) and 15.1 versus 14.4 months (P = 0.48) in cases with and without statin use. The use of statin was not associated with PFS (hazard ratio, 1.01; 95% confidence interval, 0.85-1.18, P = 0.93) or OS (hazard ratio, 1.05; 95% confidence interval, 0.91-1.21, P = 0.47) in the multivariable analyses. PFS and OS did not significantly differ by liposolubility of statins, either. CONCLUSIONS Stain use was not associated with PFS or OS in patients with unresectable PC receiving GnP.
Collapse
Affiliation(s)
| | | | - Takashi Sasaki
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tsuyoshi Takeda
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Shun Tezuka
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Hiroyuki Isayama
- Department of Gastroenterology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Ko Tomishima
- Department of Gastroenterology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yasushi Kojima
- Department of Gastroenterology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Natsuyo Yamamoto
- Department of Gastroenterology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yukiko Ito
- Department of Gastroenterology, Japanese Red Cross Medical Center, Tokyo, Japan
| | | | - Nobuo Toda
- Department of Gastroenterology, Mitsui Memorial Hospital, Tokyo, Japan
| | - Kaoru Takagi
- Department of Gastroenterology, Mitsui Memorial Hospital, Tokyo, Japan
| | - Saburo Matsubara
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Dai Mohri
- Department of Gastroenterology, JR Tokyo general hospital, Tokyo, Japan
| | | | - Mitsuhiro Fujishiro
- From the Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
116
|
Brown MB, Blair HA. Liposomal Irinotecan: A Review as First-Line Therapy in Metastatic Pancreatic Adenocarcinoma. Drugs 2025; 85:255-262. [PMID: 39820839 DOI: 10.1007/s40265-024-02133-1] [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: 11/21/2024] [Indexed: 01/19/2025]
Abstract
Liposomal irinotecan (Onivyde®), also known as liposomal pegylated irinotecan, has been developed with the intent of maximising anti-tumour efficacy and minimising drug-related toxicities compared with conventional formulations of this topoisomerase 1 inhibitor. In combination with fluorouracil, leucovorin and oxaliplatin (NALIRIFOX), liposomal irinotecan is approved in the USA and the EU for first-line therapy of eligible patients with metastatic pancreatic adenocarcinoma. In a phase III clinical trial, NALIRIFOX significantly improved overall survival (OS) and progression free survival (PFS) compared with gemcitabine plus nanoparticle albumin bound paclitaxel (nab-paclitaxel) as first-line treatment of patients with metastatic pancreatic ductal adenocarcinoma (mPDAC). The safety profile of NALIRIFOX was generally manageable, with diarrhoea, hypokalaemia and neutropenia being the most common grade ≥ 3 treatment-emergent adverse events. Although further analyses will help position the liposomal irinotecan-containing regimen NALIRIFOX in first-line treatment of metastatic pancreatic adenocarcinoma, current evidence indicates that it is a useful addition to treatment options in this patient population.
Collapse
Affiliation(s)
- Michael B Brown
- Springer Nature, Mairangi Bay, Private Bag 65901, Auckland, 0754, New Zealand.
| | - Hannah A Blair
- Springer Nature, Mairangi Bay, Private Bag 65901, Auckland, 0754, New Zealand
| |
Collapse
|
117
|
Bruciamacchie M, Garambois V, Vie N, Bessede T, Michaud HA, Chepeaux LA, Gros L, Bonnefoy N, Robin M, Brager D, Bigot K, Evrard A, Pourquier P, Colinge J, Mathonnet M, Belhabib I, Jean C, Bousquet C, Colombo PE, Jarlier M, Tosi D, Gongora C, Larbouret C. ATR inhibition potentiates FOLFIRINOX cytotoxic effect in models of pancreatic ductal adenocarcinoma by remodelling the tumour microenvironment. Br J Cancer 2025; 132:222-235. [PMID: 39613844 PMCID: PMC11746931 DOI: 10.1038/s41416-024-02904-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 10/18/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND In pancreatic ductal adenocarcinoma (PDAC), the dense stroma rich in cancer-associated fibroblasts (CAFs) and the immunosuppressive microenvironment confer resistance to treatments. To overcome such resistance, we tested the combination of FOLFIRINOX (DNA damage-inducing chemotherapy drugs) with VE-822 (an ataxia-telangiectasia and RAD3-related inhibitor that targets DNA damage repair). METHODS PDAC spheroid models and organoids were used to assess the combination effects. Tumour growth and the immune and fibrotic microenvironment were evaluated by immunohistochemistry, single-cell analysis and spatial proteomics in patient-derived xenograft (PDX) and orthotopic immunocompetent KPC mouse models. RESULTS The FOLFIRINOX and VE-822 combination had a strong synergistic effect in several PDAC cell lines, whatever their BRCA1, BRCA2 and ATM mutation status and resistance to standard chemotherapy agents. This was associated with high DNA damage and inhibition of DNA repair signalling pathways, leading to increased apoptosis. In immunocompetent and PDX mouse models of PDAC, the combination inhibited tumour growth more effectively than FOLFIRINOX alone. This was associated with tumour microenvironment remodelling, particularly decreased proportion of fibroblast activated protein-positive CAFs and increased anti-tumorigenic immune cell infiltration and interaction. CONCLUSION The FOLFIRINOX and VE-822 combination is a promising strategy to improve FOLFIRINOX efficacy and overcome drug resistance in PDAC.
Collapse
Affiliation(s)
| | | | - Nadia Vie
- IRCM, Univ Montpellier, Inserm, ICM, Montpellier, France
| | - Thomas Bessede
- IRCM, Univ Montpellier, Inserm, ICM, Montpellier, France
| | | | | | - Laurent Gros
- IRCM, Univ Montpellier, Inserm, ICM, Montpellier, France
| | | | - Mathilde Robin
- IRCM, Univ Montpellier, Inserm, ICM, Montpellier, France
| | - Dorian Brager
- IRCM, Univ Montpellier, Inserm, ICM, Montpellier, France
| | - Kevin Bigot
- IRCM, Univ Montpellier, Inserm, ICM, Montpellier, France
| | - Alexandre Evrard
- IRCM, Univ Montpellier, Inserm, ICM, CHU Nimes, Montpellier, France
| | | | | | | | - Ismahane Belhabib
- Université Toulouse III-Paul Sabatier-Centre de Recherche en Cancérologie de Toulouse (CRCT)-UMR1037 Inserm- UMR 5071 CNRS, Toulouse, France
| | - Christine Jean
- Université Toulouse III-Paul Sabatier-Centre de Recherche en Cancérologie de Toulouse (CRCT)-UMR1037 Inserm- UMR 5071 CNRS, Toulouse, France
| | - Corinne Bousquet
- Université Toulouse III-Paul Sabatier-Centre de Recherche en Cancérologie de Toulouse (CRCT)-UMR1037 Inserm- UMR 5071 CNRS, Toulouse, France
| | | | - Marta Jarlier
- IRCM, Univ Montpellier, Inserm, ICM, Montpellier, France
| | - Diégo Tosi
- IRCM, Univ Montpellier, Inserm, ICM, Montpellier, France
| | - Céline Gongora
- IRCM, Univ Montpellier, Inserm, ICM, CNRS, Montpellier, France
| | | |
Collapse
|
118
|
Miyazaki S, Kitazawa M, Nakamura S, Koyama M, Yamamoto Y, Hondo N, Kataoka M, Tanaka H, Takeoka M, Komatsu D, Soejima Y. Targeting KRAS-mutant pancreatic cancer through simultaneous inhibition of KRAS, MEK, and JAK2. Mol Oncol 2025; 19:377-390. [PMID: 39400496 PMCID: PMC11793007 DOI: 10.1002/1878-0261.13751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 08/02/2024] [Accepted: 10/03/2024] [Indexed: 10/15/2024] Open
Abstract
The Kirsten rat sarcoma (KRAS) oncogene was considered "undruggable" until the development of sotorasib, a KRASG12C selective inhibitor that shows favorable effects against lung cancers. MRTX1133, a novel KRASG12D inhibitor, has shown promising results in basic research, although its effects against pancreatic cancer are limited when used alone. Therefore, there is an urgent need to identify effective drugs that can be used in combination with KRAS inhibitors. In this study, we found that administration of the KRAS inhibitors sotorasib or MRTX1133 upregulated STAT3 phosphorylation and reactivated ERK through a feedback reaction. The addition of the MEK inhibitor trametinib and the JAK2 inhibitor fedratinib successfully reversed this effect and resulted in significant growth inhibition in vitro and in vivo. Analyses of sotorasib- and MRTX1133-resistant cells showed that trametinib plus fedratinib reversed the resistance to sotorasib or MRTX1133. These findings suggest that the JAK2-mediated pathway and reactivation of the MAPK pathway may play key roles in resistance to KRAS inhibitors in pancreatic cancers. Accordingly, simultaneous inhibition of KRAS, MEK, and JAK2 could be an innovative therapeutic strategy against KRAS-mutant pancreatic cancer.
Collapse
Affiliation(s)
- Satoru Miyazaki
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Masato Kitazawa
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Satoshi Nakamura
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Makoto Koyama
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Yuta Yamamoto
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Nao Hondo
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Masahiro Kataoka
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Hirokazu Tanaka
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Michiko Takeoka
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | | | - Yuji Soejima
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| |
Collapse
|
119
|
Ishigaki K, Tokito Y, Takahara N, Nishio H, Endo G, Fukuda K, Ishida K, Fukuda R, Takaoka S, Oyama H, Noguchi K, Suzuki T, Sato T, Saito T, Hamada T, Miyabayashi K, Sato Y, Nakai Y, Kage H, Oda K, Fujishiro M. Association between homologous recombination deficiency and time to treatment failure to platinum-based chemotherapy for pancreatic cancer by using the C-CAT database. J Gastroenterol 2025; 60:247-256. [PMID: 39570378 PMCID: PMC11794350 DOI: 10.1007/s00535-024-02173-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 11/03/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Since homologous recombination deficiency (HRD) is relatively uncommon in pancreatic cancer (PC), its impact on time-to-treatment failure (TTF) among patients undergoing systemic chemotherapy for unresectable and recurrent PC remains uncertain. METHODS Among patients with unresectable and recurrent PC enrolled in the Center for Cancer Genomics and Advanced Therapeutics (C-CAT) database by July 2023, a total of 1394 patients who underwent first-line chemotherapy with either gemcitabine plus nab-paclitaxel (GnP) or FOLFIRINOX (FFX) and received tissue-based CGP tests after disease progression were included in this study. HRD was defined as the presence of germline or somatic genetic mutations in homologous recombination repair (HRR)-related genes such as ATM, BARD1, BRIP1, BRCA1/2, CHEK2, CDK12, PALB, and RAD51C/D. We investigated the correlation between HRD and TTF among patients treated with GnP and FFX. RESULTS First-line chemotherapy consisted of GnP in 69% of the cases and FFX in 31%. The CGP tests used were NCC OncoPanel and FoundationOne CDx in 26% and 74%, respectively. HRR-related genetic abnormalities were identified in 107 patients (7.6%): BRCA2 (n = 51), ATM (n = 34), BRCA1 (n = 9), PALB2 (n = 9), among others. In the GnP cohort, the median TTF was comparable between the HRD and non-HRD groups (5.3 vs 4.6 months, P = 0.44). Conversely, in the FFX cohort, it was significantly longer in the HRD group compared to the non-HRD group (7.3 vs. 4.7 months, p < 0.01). CONCLUSIONS Our findings suggest that HRR-related genetic abnormalities might be predictive of TTF in platinum-based chemotherapy for PC.
Collapse
Affiliation(s)
- Kazunaga Ishigaki
- Department of Clinical Oncology, The University of Tokyo Hospital, Tokyo, Japan
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Yurie Tokito
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Naminatsu Takahara
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan.
| | - Hiroto Nishio
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Go Endo
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Koshiro Fukuda
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Kota Ishida
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Rintaro Fukuda
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Shinya Takaoka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Hiroki Oyama
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Kensaku Noguchi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Tatsunori Suzuki
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Tatsuya Sato
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Tomotaka Saito
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Tsuyoshi Hamada
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Koji Miyabayashi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Yasuyoshi Sato
- Department of Clinical Oncology, The University of Tokyo Hospital, Tokyo, Japan
| | - Yousuke Nakai
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
- Department of Internal Medicine, Institute of Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - Hidenori Kage
- Department of Respiratory Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Katsutoshi Oda
- Department of Clinical Genomics, The University of Tokyo Hospital, Tokyo, Japan
| | - Mitsuhiro Fujishiro
- Department of Clinical Oncology, The University of Tokyo Hospital, Tokyo, Japan
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-Ku, Tokyo, 113-8655, Japan
| |
Collapse
|
120
|
Amrutkar M, Guttorm SJT, Finstadsveen AV, Labori KJ, Eide L, Rootwelt H, Elgstøen KBP, Gladhaug IP, Verbeke CS. Global metabolomic profiling of tumor tissue and paired serum samples to identify biomarkers for response to neoadjuvant FOLFIRINOX treatment of human pancreatic cancer. Mol Oncol 2025; 19:391-411. [PMID: 39545923 PMCID: PMC11793008 DOI: 10.1002/1878-0261.13759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/26/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024] Open
Abstract
Neoadjuvant chemotherapy (NAT) is increasingly used for the treatment of non-metastatic pancreatic ductal adenocarcinoma (PDAC) and is established as a standard of care for borderline resectable and locally advanced PDAC. However, full exploitation of its clinical benefits is limited by the lack of biomarkers that assess treatment response. To address this unmet need, global metabolomic profiling was performed on tumor tissue and paired serum samples from patients with treatment-naïve (TN; n = 18) and neoadjuvant leucovorin calcium (folinic acid), fluorouracil, irinotecan hydrochloride and oxaliplatin (FOLFIRINOX)-treated (NAT; n = 17) PDAC using liquid chromatography mass spectrometry. Differentially abundant metabolites (DAMs) in TN versus NAT groups were identified and their correlation with various clinical parameters was assessed. Metabolomics profiling identified 40 tissue and five serum DAMs in TN versus NAT PDAC. In general, DAMs associated with amino acid and nucleotide metabolism were lower in NAT compared to TN. Four DAMs-3-hydroxybutyric acid (BHB), 3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid (CMPF), glycochenodeoxycholate and citrulline-were common to both tissue and serum and showed a similar pattern of differential abundance in both groups. A strong positive correlation was observed between serum carbohydrate 19-9 antigen (CA 19-9) and tissue carnitines (C12, C18, C18:2) and N8-acetylspermidine. The reduction in CA 19-9 following NAT correlated negatively with serum deoxycholate levels, and the latter correlated positively with survival. This study revealed neoadjuvant-chemotherapy-induced changes in metabolic pathways in PDAC, mainly amino acid and nucleotide metabolism, and these correlated with reduced CA 19-9 following neoadjuvant FOLFIRINOX treatment.
Collapse
Affiliation(s)
- Manoj Amrutkar
- Department of Pathology, Division of Laboratory MedicineOslo University HospitalNorway
| | - Sander Johannes Thorbjørnsen Guttorm
- Department of Medical Biochemistry, Division of Laboratory MedicineOslo University HospitalNorway
- Core Facility for Global Metabolomics and Lipidomics, Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloNorway
| | | | - Knut Jørgen Labori
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloNorway
- Department of Hepato‐Pancreato‐Biliary SurgeryOslo University HospitalOsloNorway
| | - Lars Eide
- Department of Medical Biochemistry, Division of Laboratory MedicineOslo University HospitalNorway
- Department of Medical Biochemistry, Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloNorway
| | - Helge Rootwelt
- Department of Medical Biochemistry, Division of Laboratory MedicineOslo University HospitalNorway
- Core Facility for Global Metabolomics and Lipidomics, Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloNorway
| | - Katja Benedikte Prestø Elgstøen
- Department of Medical Biochemistry, Division of Laboratory MedicineOslo University HospitalNorway
- Core Facility for Global Metabolomics and Lipidomics, Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloNorway
| | - Ivar P. Gladhaug
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloNorway
- Department of Hepato‐Pancreato‐Biliary SurgeryOslo University HospitalOsloNorway
| | - Caroline S. Verbeke
- Department of Pathology, Division of Laboratory MedicineOslo University HospitalNorway
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloNorway
| |
Collapse
|
121
|
Min JH, Yu JI, Kim SH, Kim YK, Kim K, Park HC, Park JO, Hong JY, Lee KT, Lee KH, Lee JK, Park JK, Choi JH, Heo JS, Han IW, Kim H, Shin SH, Yoon SJ, Woo SY. Skeletal Muscle Index Changes on Locoregional Treatment Application After FOLFIRINOX and Survival in Pancreatic Cancer. J Cachexia Sarcopenia Muscle 2025; 16:e16343. [PMID: 39578950 DOI: 10.1002/jcsm.13643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Patients with borderline resectable (BR) or locally advanced pancreatic cancer (LAPC) require complex management strategies. This study evaluated the prognostic significance of the perichemotherapy skeletal muscle index (SMI) and carbohydrate antigen 19-9 (CA 19-9) in patients with BRPC or LAPC treated with FOLFIRINOX. METHODS We retrospectively evaluated 227 patients with BR or LAPC who received at least four cycles of chemotherapy between 2015 and 2020. We analysed chemotherapy response, changes in SMI (ΔSMI, %) on computed tomography (CT) and CA19-9 to determine their impact on progression-free survival (PFS) and overall survival (OS). After the early application of loco-regional treatments (LRT) within 3 months after completing four cycles of chemotherapy, the outcomes were compared between ΔSMI and CA19-9 subgroups. RESULTS Among 227 patients (median age, 60 years; 124 [54.6%] male) with 97 BR and 130 LAPC, 50.7% showed partial response (PR) to chemotherapy, 44.5% showed stable disease and 4.8% showed progressive disease (PD). Post-chemotherapy CA19-9 levels were normalized in 41.0% of patients. The high and low ΔSMI groups (based on the gender-specific cut-off of -8.6% for males and -2.9% for females) comprised 114 (50.2%) and 113 (49.8%) patients, respectively. The high ΔSMI group had poorer survival rates than the low ΔSMI group in both PFS (HR = 1.32, p = 0.05) and OS (HR = 1.74, p = 0.001). Multivariable analysis showed that ΔSMI (high vs. low; PFS, HR = 1.39, p = 0.03; OS, HR = 1.82, p < 0.001) and post-chemotherapy response (PD vs. PR/SD; PFS, HR = 18.69, p < 0.001; OS, HR = 6.19, p < 0.001) were independently associated with both PFS and OS. Additionally, the post-chemotherapy CA19-9 (≥ 37 vs. < 37; HR = 1.48, p = 0.01) was an independent predictor for PFS. Early application of LRT after chemotherapy significantly improved PFS and OS in both ΔSMI groups (all p < 0.05). However, it was not beneficial in the group with high ΔSMI and post-chemotherapy CA19-9 ≥ 37 (PFS, p = 0.39 and OS, p = 0.33). CONCLUSIONS Progressive sarcopenic deterioration after four cycles of chemotherapy was associated with poor survival outcomes in patients with BR or LAPC after FOLFIRINOX. We also investigated the optimal clinical setting for the early application LRTs using the ΔSMI and post-chemotherapy CA 19-9.
Collapse
Affiliation(s)
- Ji Hye Min
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jeong Il Yu
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Seong Hyun Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Young Kon Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kangpyo Kim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hee Chul Park
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Joon Oh Park
- Divisions of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jung Yong Hong
- Divisions of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyu Taek Lee
- Divisions of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kwang Hyuck Lee
- Divisions of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jong Kyun Lee
- Divisions of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Joo Kyung Park
- Divisions of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jin Ho Choi
- Divisions of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jin Seok Heo
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - In Woong Han
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hongbeom Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sang Hyun Shin
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - So Jung Yoon
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sook-Young Woo
- Biomedical Statistics Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea
| |
Collapse
|
122
|
Liu R, Ji Z, Wang X, Xin J, Zhu L, Ge S, Zhang L, Bai M, Ning T, Yang Y, Li H, Deng T, Ba Y. Efficacy and safety of multi-target tyrosine kinase inhibitor AL2846 combined with gemcitabine in pancreatic cancer. Invest New Drugs 2025; 43:81-92. [PMID: 39760815 DOI: 10.1007/s10637-024-01485-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/06/2024] [Indexed: 01/07/2025]
Abstract
Pancreatic cancer patients urgently need new treatments, and we explored the efficacy and safety of combination therapy with AL2846 and gemcitabine in pancreatic cancer patients. This was a single-arm, single-center, open-label phase I/IIa study (NCT06278493). The dose-escalation phase was designed to evaluate the maximum tolerated dose (MTD) of AL2846 combined with gemcitabine. One or two dose levels were chosen for the dose-expansion phase. Treatment continued until disease progression, intolerable toxicity, patient withdrawal, or at the investigators' discretion. The primary study endpoint is to evaluate the safety and MTD of AL2846 combined with gemcitabine. The secondary endpoints included objective response rate (ORR), progression-free survival (PFS), overall survival (OS), and disease control rate (DCR). Between August 2018 and July 2021, 33 pancreatic cancer patients were enrolled in the study. A total of 15 patients were enrolled in the dose-escalation phase, and the MTD was not determined. Eventually 90 mg and 120 mg of AL2846 were chosen for the dose-expansion phase, in which 11 patients (90 mg) and 7 patients (120 mg) were administered. Treatment-related adverse events (TRAEs) of any grade were reported in 30 (90.91%) patients, and those of grade ≥ 3 were reported in 16 (48.48%) patients. The most frequently reported grade ≥ 3 TRAEs were thrombocytopenia (18.18%), neutropenia (12.12%), elevated γ-glutamyltransferase (6.06%), proteinuria (6.06%), and gastrointestinal hemorrhage (6.06%).The ORR was 6.06%, and the DCR was 72.73%. The median PFS was 3.71 months (95% CI: 3.38-4.11), and the median OS was 5.59 months (95% CI: 4.11-8.71). Gemcitabine and Al2846 combination therapy exhibited tolerable safety, but there was no improvement in efficacy over standard treatment. Further evaluation of this approach is still needed.
Collapse
Affiliation(s)
- Rui Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zhi Ji
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xia Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jiaqi Xin
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Lila Zhu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Shaohua Ge
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Le Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Ming Bai
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Tao Ning
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yuchong Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Hongli Li
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| | - Ting Deng
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| | - Yi Ba
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
123
|
Yamaguchi T, Kitahara S, Matsui A, Okamoto J, Muragaki Y, Masamune K. HIFU induces reprogramming of the tumor immune microenvironment in a pancreatic cancer mouse model. Med Mol Morphol 2025:10.1007/s00795-025-00419-1. [PMID: 39870899 DOI: 10.1007/s00795-025-00419-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/06/2025] [Indexed: 01/29/2025]
Abstract
This study evaluates the effects of different high-intensity focused ultrasound irradiation (HIFU) methods on local tumor suppression and systemic antitumor effects, including the abscopal effect, in a mouse model of pancreatic cancer. To ascertain the efficacy of the treatment, pancreatic cancer cells were injected into the thighs of mice and HIFU was applied on one side using continuous waves or trigger pulse waves. Then, tumor volume, tissue changes, and immune marker levels were analyzed. Both the irradiation methods suppressed tumor growth, with the trigger pulse wave showing stronger effects and the difference being significant. Tumor suppression was also observed on the non-irradiated side, suggesting an abscopal effect. These effects vary depending on the irradiation method used. We conclude that HIFU induces both local tumor suppression and a systemic immune response, suggesting its potential for combination with immunotherapy for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Toshihiro Yamaguchi
- Faculty of Advanced Techno-Surgery (FATS), Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan
| | - Shuji Kitahara
- Faculty of Advanced Techno-Surgery (FATS), Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan.
| | - Aya Matsui
- Department of Vascular Physiology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Jun Okamoto
- SONIRE Therapeutics Inc., Nihonbashi Life Science, Building 2 803, 3-11-5 Nihonbashi Honcho, Chuo-Ku 103-0023, Tokyo, Japan
| | - Yoshihiro Muragaki
- Faculty of Advanced Techno-Surgery (FATS), Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan
- Center for Advanced Medical Engineering Research and Development, Kobe University, 1-5-1 Minatojima Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Ken Masamune
- Faculty of Advanced Techno-Surgery (FATS), Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan.
| |
Collapse
|
124
|
Maekawa A, Omiya K, Oba A, Inoue Y, Hirose Y, Kobayashi K, Ono Y, Sato T, Sasaki T, Ozaka M, Matsueda K, Mise Y, Takamatsu M, Shigematsu Y, Ito H, Saiura A, Sasahira N, Takahashi Y. Clinical implications of disappearing pancreatic cancer liver metastases: Lessons from colorectal liver metastases. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109635. [PMID: 39879814 DOI: 10.1016/j.ejso.2025.109635] [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: 11/05/2024] [Revised: 12/27/2024] [Accepted: 01/23/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND The efficacy of local control for pancreatic cancer liver metastases (PCLM), including surgical treatment, remains controversial, with no consensus on the management and clinical significance of disappearing liver metastases (DLMs). This study aimed to evaluate the clinical implications of DLMs in treating PCLM after multi-agent chemotherapy, utilizing contrast-enhanced imaging modalities. METHODS A retrospective analysis was conducted on patients who underwent curative resection for pancreatic cancer with synchronous or metachronous liver metastases between 2014 and 2023. Surgical indications were based on our recently reported ABCD criteria (Anatomical/Biological/Conditional/Duration). Both contrast-enhanced computed tomography (CE-CT) and gadolinium ethoxybenzyl diethylenetriamine pentaacetic acid-enhanced magnetic resonance imaging (EOB-MRI) were used to monitor metastatic lesions in the liver. DLMs were defined as tumors undetected on CE-CT post-chemotherapy. RESULTS A total of 58 lesions in 29 patients with PCLM who underwent surgical resection were evaluated. Of the 13 lesions evident on CE-CT, 76.9 % (10/13) contained clinically/pathologically residual tumors. Of the 45 DLMs, 16 (35.6 %) had residual tumors. Twenty-six DLMs (57.8 %) were detected on EOB-MRI or intraoperative screening (contrast-enhanced ultrasonography and palpation), with 42.3 % (11/26) being residual tumors. Nineteen DLMs were undetectable by any modality, of which 26.3 % (5/19) were confirmed to be residual tumors with a median follow-up of 32 months. The median overall survival from initiating treatment for PCLM was 48.5 months. CONCLUSION Integrating EOB-MRI into preoperative evaluations for PCLM enhances the detection of clinically relevant DLMs. Our findings highlight the potential benefits of considering an image-guided surgical approach in selected patients.
Collapse
Affiliation(s)
- Aya Maekawa
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kojiro Omiya
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Atsushi Oba
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Yosuke Inoue
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yuki Hirose
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan; Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kosuke Kobayashi
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoshihiro Ono
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takafumi Sato
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takashi Sasaki
- Department of Gastroenterological Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masato Ozaka
- Department of Gastroenterological Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kiyoshi Matsueda
- Department of Diagnostic Imaging, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoshihiro Mise
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Manabu Takamatsu
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yasuyuki Shigematsu
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiromichi Ito
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akio Saiura
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Naoki Sasahira
- Department of Gastroenterological Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yu Takahashi
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.
| |
Collapse
|
125
|
Araki T, Sonoda Y, Shimokawa M, Otsuka T, Hayashi K, Honda T, Nakao K, Shibuki T, Nakazawa J, Arima S, Miwa K, Okabe Y, Koga F, Ueda Y, Kubotsu Y, Shimokawa H, Takeshita S, Komori A, Nishikawa K, Otsu S, Hosokawa A, Oda H, Sakai T, Arita S, Kawahira M, Taguchi H, Tsuneyoshi K, Kawaguchi Y, Fujita T, Sakae T, Shirakawa T, Mizuta T, Mitsugi K. Relationship between neutropenia caused by nanoliposomal irinotecan/fluorouracil/leucovorin and treatment outcomes in the NAPOLEON-2 study (NN-2301). Sci Rep 2025; 15:3427. [PMID: 39870769 PMCID: PMC11772893 DOI: 10.1038/s41598-025-88005-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/23/2025] [Indexed: 01/29/2025] Open
Abstract
The relationship between nanoliposomal irinotecan/fluorouracil/leucovorin (NFF) treatment outcomes and neutropenia in patients with pancreatic cancer has not been thoroughly examined. Thus, we conducted a retrospective analysis of data from patients with pancreatic cancer who were treated with NFF to investigate this relationship. Neutropenia was assessed according to the Common Terminology Criteria for Adverse Events across three cutoffs: A (grade 0 versus grade 1-4), B (grades 0-1 versus 2-4), and C (grades 0-2 versus 3-4). The primary endpoint was overall survival (OS), and the secondary endpoints were overall response rate, progression-free survival (PFS), and relative dose intensity. Of the 161 patients, 93, 8, 22, 30, and 8 patients had neutropenia of grades 0, 1, 2, 3, and 4, respectively. The overall response rates differed significantly at cutoff C (p = 0.02), with the odds ratio for cutoff C being the highest, followed by cutoffs B and A. Significant differences in OS were observed at cutoffs A (hazard ratio [HR], 0.65; 95% confidence interval [CI], 0.44-0.94; p = 0.02) and B (HR, 0.63; 95% CI, 0.43-0.92, p = 0.02). Similarly, PFS showed significant differences at cutoffs A and B (p < 0.01). NFF-induced neutropenia can be a useful prognostic factor for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Tomonori Araki
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yuki Sonoda
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Mototsugu Shimokawa
- Clinical Research Institute, National Kyushu Cancer Center, Fukuoka, Japan
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Taiga Otsuka
- Department of Internal Medicine, Minato Medical Clinic, Fukuoka, Japan
| | - Kohei Hayashi
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Taro Shibuki
- Department for the Promotion of Drug and Diagnostic Development, Division of Drug and Diagnostic Development Promotion, Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Junichi Nakazawa
- Department of Medical Oncology, Kagoshima City Hospital, Kagoshima, Japan
| | - Shiho Arima
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Keisuke Miwa
- Multidisciplinary Treatment Cancer Center, Kurume University Hospital, Fukuoka, Japan
| | - Yoshinobu Okabe
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Futa Koga
- Department of Hepatobiliary and Pancreatology, Saga Medical Center Koseikan, Saga, Japan
| | - Yujiro Ueda
- Department of Hematology and Oncology, Japanese Red Cross Kumamoto Hospital, Kumamoto, Japan
| | - Yoshihito Kubotsu
- Department of Internal Medicine, Karatsu Red Cross Hospital, Saga, Japan
| | - Hozumi Shimokawa
- Department of Hematology Oncology, Community Healthcare Organization Kyushu Hospital, Fukuoka, Japan
| | - Shigeyuki Takeshita
- Department of Gastroenterology, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Azusa Komori
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Oita, Japan
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Ehime, Japan
| | - Kazuo Nishikawa
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Oita, Japan
| | - Satoshi Otsu
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Oita, Japan
| | - Ayumu Hosokawa
- Department of Clinical Oncology, University of Miyazaki Hospital, Miyazaki, Japan
| | - Hisanobu Oda
- Division of Integrative Medical Oncology, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Tatsunori Sakai
- Department of Medical Oncology, NHO Kumamoto Medical Center, Kumamoto, Japan
| | - Shuji Arita
- Department of Chemotherapy, Miyazaki Prefectural Miyazaki Hospital, Miyazaki, Japan
| | - Machiko Kawahira
- Department of Gastroenterology, Kagoshima Kouseiren Hospital, Kagoshima, Japan
| | - Hiroki Taguchi
- Department of Gastroenterology, Izumi General Medical Center, Kagoshima, Japan
- Department of Gastroenterology, Kagoshima City Hospital, Kagoshima, Japan
| | - Kengo Tsuneyoshi
- Department of Gastroenterology, Izumi General Medical Center, Kagoshima, Japan
| | - Yasunori Kawaguchi
- Department of Gastroenterology, Asakura Medical Association Hospital, Fukuoka, Japan
| | - Toshihiro Fujita
- Department of Gastroenterology, Saiseikai Sendai Hospital, Kagoshima, Japan
| | - Takahiro Sakae
- Department of Gastroenterology, Saiseikai Sendai Hospital, Kagoshima, Japan
| | - Tsuyoshi Shirakawa
- Researcher of Clinical Hematology Oncology Treatment Study Group, 1-14-6 Muromi-gaoka, Nishi-ku, Fukuoka-shi, Fukuoka, 819-0030, Japan.
- Director of Medical Checkup Center, Eikoh Hospital, 3-8-15 Befu-nishi, Shime-machi, Kasuya- gun, Fukuoka, 811-2232, Japan.
| | | | - Kenji Mitsugi
- Department of Medical Oncology, Sasebo Kyosai Hospital, Sasebo, Japan
| |
Collapse
|
126
|
Chang E, Sherry AD, Liermann J, Abdollahi A, Tzeng CWD, Tang C, Aguilera TA, Koay EJ, Das P, Koong AC, Pant S, Ludmir EB. Evolving Paradigms in the Treatment of Oligometastatic Pancreatic Ductal Adenocarcinoma. J Gastrointest Cancer 2025; 56:47. [PMID: 39827280 DOI: 10.1007/s12029-024-01145-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2024] [Indexed: 01/22/2025]
Abstract
Multiple randomized trials have suggested that the addition of comprehensive metastasis-directed therapy to best systemic therapy improves disease control and survival among patients with oligometastatic disease, even for histologies with a high propensity for rapid spread. Here, we review the growing literature supporting the oligometastatic paradigm in pancreatic ductal adenocarcinoma. We summarize key details from nascent institutional series and reflect on the recently reported phase II randomized EXTEND trial. We discuss various strategies for enhancing the clinical and technical implementation of metastasis-directed therapy in this patient population. Lastly, we highlight multiple ongoing landmark trials seeking to optimize and validate the role of metastasis-directed therapy in oligometastatic pancreatic cancer. Ultimately, these and other continued clinical and translational research efforts will be critical to improve care and outcomes for patients with oligometastatic pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Enoch Chang
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander D Sherry
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jakob Liermann
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Amir Abdollahi
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Ching-Wei D Tzeng
- Department of Surgical Oncology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chad Tang
- Department of Genitourinary Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Todd A Aguilera
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eugene J Koay
- Department of Gastrointestinal Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prajnan Das
- Department of Gastrointestinal Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Albert C Koong
- Department of Gastrointestinal Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ethan B Ludmir
- Department of Gastrointestinal Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
127
|
Zhang Z, Xu H, He J, Hu Q, Liu Y, Xu Z, Lou W, Wu W, Zhang L, Pu N, Shi C, Xu Y, Wang W, Liu L. Inhibition of KLF5 promotes ferroptosis via the ZEB1/HMOX1 axis to enhance sensitivity to oxaliplatin in cancer cells. Cell Death Dis 2025; 16:28. [PMID: 39827156 PMCID: PMC11743205 DOI: 10.1038/s41419-025-07330-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 12/04/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
As a novel form of nonapoptotic cell death, ferroptosis is developing into a promising therapeutic target of dedifferentiating and therapy-refractory cancers. However, its application in pancreatic cancer is still unknown. In the preliminary research, we found that F-box and WD repeat domain-containing 7 (FBW7) inhibited the migration and proliferation of pancreatic cancer cells through its substrate c-Myc. We further found that another key substrate of FBW7, KLF5, could inhibit ferroptosis. Inhibiting KLF5 significantly enhances the cytotoxicity of oxaliplatin rather than other chemotherapy drugs. Mechanistically, we found that KLF5 inhibited the expression of heme oxygenase 1 (HMOX1) via repressing zinc finger E-box-binding homeobox 1 (ZEB1). Inhibition of KLF5 facilitated the cytotoxic effect of oxaliplatin via promoting ferroptosis. Oxaliplatin combined with KLF5 inhibitor significantly potentiated cell death in vitro and inhibited tumor growth in vivo compared with either treatment alone. These results reveal a critical role of KLF5 in sensitized chemotherapy of pancreatic cancer, and suggest that ferroptosis combined with platinum-based chemotherapy rather than gemcitabine-based chemotherapy is expected to bring better therapeutic effects.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huaxiang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junyi He
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiangsheng Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuxin Liu
- Institute of liver diseases, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zijin Xu
- Department of General Surgery, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Wenhui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenchuan Wu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lei Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ning Pu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chenye Shi
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yaolin Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
128
|
Sarno F, Tenorio J, Perea S, Medina L, Pazo-Cid R, Juez I, Garcia-Carbonero R, Feliu J, Guillen-Ponce C, Lopez-Casas PP, Guerra C, Duran Y, López-Acosta JF, Alonso C, Esquivel E, Dopazo A, Akshinthala D, Muthuswamy SK, Lapunzina P, Bockorny B, Hidalgo M. A Phase III Randomized Trial of Integrated Genomics and Avatar Models for Personalized Treatment of Pancreatic Cancer: The AVATAR Trial. Clin Cancer Res 2025; 31:278-287. [PMID: 39540844 PMCID: PMC11739777 DOI: 10.1158/1078-0432.ccr-23-4026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/24/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) has limited treatment options. We compared the efficacy of comprehensive precision medicine against that of the conventional treatment in PDAC. PATIENTS AND METHODS We report a phase III trial of advanced PDAC in which patients were randomized (1:2) to a conventional treatment treated at physician's discretion (arm A) or to precision medicine (arm B). Subjects randomized to arm B underwent a tumor biopsy for whole-exome sequencing and to generate avatar mouse models and patient-derived organoids for phenotypic drug screening, with final treatment recommended by the molecular tumor board. The primary objective was median overall survival (OS). RESULTS A total of 137 patients were enrolled with 125 randomized, 44 to arm A and 81 to arm B. Whole-exome sequencing was performed in 80.3% (65/81) patients of arm B, with potentially actionable mutations detected in 21.5% (14/65). Experimental models were generated in 16/81 patients (19.8%). Second-line treatment was administered to 39 patients in the experimental arm, but only four (10.2%) received personalized treatment, whereas 35 could not receive matched therapy because of rapid clinical deterioration, delays in obtaining study results, or the absence of actionable targets. The median OS was 8.7 and 8.6 months (P = 0.849) and the median progression-free survival was 3.8 and 4.3 months (P = 0.563) for the conventional and experimental arms, respectively. Notably, the four patients who received personalized treatment had a median OS of 19.3 months. CONCLUSIONS Personalized medicine was challenging to implement in most patients with PDAC, limiting the interpretation of intention-to-treat analysis. Survival was improved in the subset of patients who did receive matched therapy.
Collapse
Affiliation(s)
| | - Jair Tenorio
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
- INGEMM-IdiPAZ Institute of Medical and Molecular Genetics, Madrid, Spain
- ITHACA, European Reference Network, Brussels, Belgium
| | - Sofia Perea
- Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Laura Medina
- UGCI Medical Oncology, Hospital Regional y Virgen de la Victoria, IBIMA, Malaga, Spain
| | | | - Ignacio Juez
- Hospital Universitario de Fuenlabrada, Madrid, Spain
| | | | - Jaime Feliu
- Hospital Universitario La Paz, Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Pedro P. Lopez-Casas
- Instituto de Investigación Sanitaria, Hospital 12 de octubre (imas 12), Madrid, Spain
| | - Carmen Guerra
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Experimental Oncology Group, Molecular Oncology Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Yolanda Duran
- Hospital Universitario de Fuenlabrada, Madrid, Spain
| | | | | | - Estrella Esquivel
- Unidad de Genómica, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ana Dopazo
- Unidad de Genómica, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Dipikaa Akshinthala
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | | | - Pablo Lapunzina
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
- INGEMM-IdiPAZ Institute of Medical and Molecular Genetics, Madrid, Spain
- ITHACA, European Reference Network, Brussels, Belgium
| | - Bruno Bockorny
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Manuel Hidalgo
- Division of Hematology-Oncology, Weill Cornell Medical College, New York, New York
- New York-Presbyterian Hospital, New York, New York
| |
Collapse
|
129
|
Sishc BJ, Saha J, Alves EM, Ding L, Lu H, Wang SY, Swancutt KL, Nicholson JH, Facoetti A, Pompos A, Ciocca M, Aguilera TA, Story MD, Davis AJ. Defective homologous recombination and genomic instability predict increased responsiveness to carbon ion radiotherapy in pancreatic cancer. NPJ Precis Oncol 2025; 9:20. [PMID: 39824957 PMCID: PMC11742413 DOI: 10.1038/s41698-025-00800-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 01/03/2025] [Indexed: 01/20/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is notably resistant to conventional chemotherapy and radiation treatment. However, clinical trials indicate that carbon ion radiotherapy (CIRT) with concurrent gemcitabine is effective for unresectable locally advanced PDAC. This study aimed to identify patient characteristics predictive of CIRT response. We utilized a panel of human PDAC cell lines with diverse genetic profiles to determine their sensitivity to CIRT compared to γ-rays, assessing relative biological effectiveness (RBE) at 10% survival, which ranged from 1.96 to 3.04. Increased radiosensitivity was linked to impaired DNA double-strand break (DSB) repair, particularly in cell lines with deficiencies in the homologous recombination (HR) repair pathway and/or elevated genomic instability from replication stress. Furthermore, pretreatment with the HR inhibitor B02 significantly enhanced CIRT sensitivity in a radioresistant PDAC cell line when irradiated in the spread-out Bragg peak but not at the entry position of the beam. These findings suggest that PDAC tumors with HR pathway mutations or high replication stress are more likely to benefit from CIRT while minimizing normal tissue toxicity.
Collapse
Affiliation(s)
- Brock J Sishc
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Mayo Clinic Florida, Jacksonville, FL, USA
| | - Janapriya Saha
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elizabeth M Alves
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lianghao Ding
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Huiming Lu
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shih-Ya Wang
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Katy L Swancutt
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - James H Nicholson
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Angelica Facoetti
- Medical Physics Unit & Research Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Arnold Pompos
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mario Ciocca
- Medical Physics Unit & Research Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Todd A Aguilera
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael D Story
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Mayo Clinic Florida, Jacksonville, FL, USA.
| | - Anthony J Davis
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
130
|
Klawitter J, Easton M, Karpeisky A, Farrell KB, Thamm DH, Shokati T, Christians U, Zinnen SP. Novel Approaches to Monitor Pharmacokinetics and Metabolism of Gemcitabine-Ibandronate Conjugate in Mice and Dogs. Molecules 2025; 30:354. [PMID: 39860223 PMCID: PMC11767451 DOI: 10.3390/molecules30020354] [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: 11/28/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND The use of the bone-seeking properties of bisphosphonates (BPs) to target the delivery of therapeutic drugs is a promising approach for the treatment of bone metastases. Currently, the most advanced example of this approach is a gemcitabine-ibandronate conjugate (GEM-IB), where the bone-targeting BP ibandronate (IB) is covalently linked to the antineoplastic agent gemcitabine (GEM) via a spacer phosphate group. In the present study, we describe the development of a new analytical platform to evaluate the metabolism and pharmacokinetics of GEM-IB in mice and dogs and the results of proof-of-concept studies assessing the pharmacokinetics of GEM-IB in dogs and mice. METHODS We validated analytical platforms to analyze GEM-IB and five of its major metabolites IB, gemcitabine-5'-phosphate (GEMMP), gemcitabine (GEM), 2',2'-difluoro-2'-deoxyuridine-5'-phosphate (dFdUMP), and 2',2'-difluoro-2'-deoxyuridine (dFdU) and performed proof-of-concept pharmacokinetic studies in mice (5 mg/kg i.p.) and dogs (5 mg/kg i.v.). RESULTS Intra- and inter-run accuracy and imprecision (3 days) of the assays met the (FDA) acceptance criteria. The proof-of-concept plasma pharmacokinetic studies in mice showed AUCs of 1278, 10,652, 405, 38, 1063, 3389, and 38 h·ng/mL for GEM-IB, IB, GEMMP, dFdU-MP, GEM, and dFdU, respectively. In dog plasma, AUCs of 295, 5725, 83, 11, 1625, and 6569 h·ng/mL were observed for GEM-IB, IB, GEMMP, dFdUMP, GEM, and dFdU. CONCLUSIONS Pharmacokinetic studies in dogs and mice showed that GEM-IB is rapidly converted to IB and GEM; dFdU is formed (from GEM) with a delay. The rapid disappearance of GEM-IB from circulation could be explained by a combination of metabolism and rapid distribution to tissue/bone.
Collapse
Affiliation(s)
- Jost Klawitter
- Department of Anesthesiology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (M.E.); (T.S.); (U.C.)
- Department of Psychiatry, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mckay Easton
- Department of Anesthesiology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (M.E.); (T.S.); (U.C.)
| | | | - Kristen B. Farrell
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO 80523, USA; (K.B.F.); (D.H.T.)
| | - Douglas H. Thamm
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO 80523, USA; (K.B.F.); (D.H.T.)
| | - Touraj Shokati
- Department of Anesthesiology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (M.E.); (T.S.); (U.C.)
| | - Uwe Christians
- Department of Anesthesiology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (M.E.); (T.S.); (U.C.)
| | | |
Collapse
|
131
|
Götze J, Meißner K, Pereira-Veiga T, Belloum Y, Schneegans S, Kropidlowski J, Gorgulho J, Busch A, Honselmann KC, Schönrock M, Putscher A, Peine S, Nitschke C, Simon R, Spindler V, Izbicki JR, Hackert T, Bokemeyer C, Pantel K, Uzunoglu FG, Sinn M, Wikman H. Identification and characterization of tumor and stromal derived liquid biopsy analytes in pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res 2025; 44:14. [PMID: 39815324 PMCID: PMC11737273 DOI: 10.1186/s13046-024-03262-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: 09/05/2024] [Accepted: 12/19/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND The lack of predictive biomarkers contributes notably to the poor outcomes of patients with pancreatic ductal adenocarcinoma (PDAC). Cancer-associated fibroblasts (CAFs) are the key components of the prominent PDAC stroma. Data on clinical relevance of CAFs entering the bloodstream, known as circulating CAFs (cCAFs) are scarce. Here, we developed a combined liquid biopsy assay to detect cCAFs and circulating tumor cells (CTCs) in metastatic PDAC (mPDAC) and other metastatic gastrointestinal malignancies (mGI). In addition, we evaluated plasma hyaluronan (HA) levels as a complementary surrogate biomarker of the stromal extent in patients with PDAC. METHODS A sequential liquid biopsy assay based on a two step-enrichment, combining marker dependent and independent cell enrichment, was established for cCAF and CTC detection and validated in mPDAC and mGI patients. The enriched cells were identified by multiplex immunofluorescence. HA measurement was performed by ELISA on blood samples from healthy blood donors (HD), localized and late-stage PDAC patients. RESULTS cCAFs (≥ 1cCAFs/7.5 mL blood) were detected in 95.4% of mPDAC and in 78.2% of mGI patients, with significantly higher numbers in mPDAC compared to mGI patients (mean number 22.7 vs. 11.0; P = 0.0318). mPDAC patients with ≥ 15 cCAFs/7.5 mL blood had a significant shorter median overall survival (mOS 3.2 months (95% confidence interval (CI) 0.801-5.855) vs. 14.2 months (95% CI 6.055-22.332); P = 0.013), whereby CTC levels were not associated with mOS. In mGI neither cCAFs nor CTCs had a significant impact on OS. HA plasma levels in mPDAC patients were significantly higher compared to HD (mean 123.0 ng/mL vs. 74.45 ng/mL, P = 0.015). High HA in localized and late-stage PDAC were associated with a significantly shorter mOS (mOSlocalized PDAC: 12.6 months vs. 23.5 months (P = 0.008); mOSmPDAC: 1.8 months vs. 5.3 months (P = 0.004)). CONCLUSIONS Our liquid biopsy assay provides robust detection of cCAFs in mPDAC and mGI patients. The measurement of both circulatory stromal parameters, cCAFs and HA, adds valuable clinical information as they are associated with an unfavorable outcome in PDAC. These results highlight that stromal characteristics unique to PDAC could be leveraged to fill the current gap in discovering predictive biomarkers.
Collapse
Affiliation(s)
- Julian Götze
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany.
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany.
| | - Kira Meißner
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Thais Pereira-Veiga
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Yassine Belloum
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Svenja Schneegans
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Jolanthe Kropidlowski
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Joao Gorgulho
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany
| | - Alina Busch
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany
| | - Kim Christin Honselmann
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Martin Schönrock
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany
| | - Arne Putscher
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Sven Peine
- Department of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Nitschke
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volker Spindler
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob Robert Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Faik Güntaç Uzunoglu
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marianne Sinn
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany.
| | - Harriet Wikman
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany.
| |
Collapse
|
132
|
Zhan T, Betge J, Schulte N, Dreikhausen L, Hirth M, Li M, Weidner P, Leipertz A, Teufel A, Ebert MP. Digestive cancers: mechanisms, therapeutics and management. Signal Transduct Target Ther 2025; 10:24. [PMID: 39809756 PMCID: PMC11733248 DOI: 10.1038/s41392-024-02097-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Cancers of the digestive system are major contributors to global cancer-associated morbidity and mortality, accounting for 35% of annual cases of cancer deaths. The etiologies, molecular features, and therapeutic management of these cancer entities are highly heterogeneous and complex. Over the last decade, genomic and functional studies have provided unprecedented insights into the biology of digestive cancers, identifying genetic drivers of tumor progression and key interaction points of tumor cells with the immune system. This knowledge is continuously translated into novel treatment concepts and targets, which are dynamically reshaping the therapeutic landscape of these tumors. In this review, we provide a concise overview of the etiology and molecular pathology of the six most common cancers of the digestive system, including esophageal, gastric, biliary tract, pancreatic, hepatocellular, and colorectal cancers. We comprehensively describe the current stage-dependent pharmacological management of these malignancies, including chemo-, targeted, and immunotherapy. For each cancer entity, we provide an overview of recent therapeutic advancements and research progress. Finally, we describe how novel insights into tumor heterogeneity and immune evasion deepen our understanding of therapy resistance and provide an outlook on innovative therapeutic strategies that will shape the future management of digestive cancers, including CAR-T cell therapy, novel antibody-drug conjugates and targeted therapies.
Collapse
Affiliation(s)
- Tianzuo Zhan
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johannes Betge
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Schulte
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Dreikhausen
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Hirth
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Moying Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philip Weidner
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Antonia Leipertz
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Teufel
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany.
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
133
|
Piotrowsky A, Burkard M, Schmieder H, Venturelli S, Renner O, Marongiu L. The therapeutic potential of vitamins A, C, and D in pancreatic cancer. Heliyon 2025; 11:e41598. [PMID: 39850424 PMCID: PMC11754517 DOI: 10.1016/j.heliyon.2024.e41598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 12/05/2024] [Accepted: 12/30/2024] [Indexed: 01/25/2025] Open
Abstract
The pancreatic ductal adenocarcinoma (PDAC) is among the deadliest tumor diseases worldwide. While treatment options have generally become more diverse, little progress has been made in the treatment of PDAC and the median survival time for patients with locally advanced PDAC is between 8.7 and 13.7 months despite treatment. The aim of this review was to explore the therapeutic potential of complementing standard therapy with natural or synthetic forms of vitamins A, C, and D. The therapeutic use of vitamins A, C, and D could be a promising addition to the treatment of PDAC. For all three vitamins and their derivatives, tumor cell-specific cytotoxicity and growth inhibition against PDAC cells has been demonstrated in vitro and in preclinical animal models. While the antitumor effect of vitamin C is probably mainly due to its pro-oxidative effect in supraphysiological concentrations, vitamin A and vitamin D exert their effect by activating nuclear receptors and influencing gene transcription. In addition, there is increasing evidence that vitamin A and vitamin D influence the tumor stroma, making the tumor tissue more accessible to other therapeutic agents. Based on these promising findings, there is a high urgency to investigate vitamins A, C, and D in a clinical context as a supplement to standard therapy in PDAC. Further studies are needed to better understand the exact mechanism of action of the individual compounds and to develop the best possible treatment regimen. This could contribute to the long-awaited progress in the treatment of this highly lethal tumor entity.
Collapse
Affiliation(s)
- Alban Piotrowsky
- Department of Nutritional Biochemistry, University of Hohenheim, 70599, Stuttgart, Germany
| | - Markus Burkard
- Department of Nutritional Biochemistry, University of Hohenheim, 70599, Stuttgart, Germany
| | - Hendrik Schmieder
- Department of Nutritional Biochemistry, University of Hohenheim, 70599, Stuttgart, Germany
| | - Sascha Venturelli
- Department of Nutritional Biochemistry, University of Hohenheim, 70599, Stuttgart, Germany
- Institute of Physiology, Department of Vegetative and Clinical Physiology, University Hospital Tuebingen, 72076, Tuebingen, Germany
| | - Olga Renner
- Department of Nutritional Biochemistry, University of Hohenheim, 70599, Stuttgart, Germany
- Faculty of Food and Nutrition Sciences, University of Applied Sciences, Hochschule Niederrhein, 41065, Moenchengladbach, Germany
| | - Luigi Marongiu
- Department of Nutritional Biochemistry, University of Hohenheim, 70599, Stuttgart, Germany
- HoLMiR-Hohenheim Center for Livestock Microbiome Research, University of Hohenheim, 70599, Stuttgart, Germany
| |
Collapse
|
134
|
Díaz-Riascos ZV, Llaguno-Munive M, Lafuente-Gómez N, Luengo Y, Holmes S, Volatron J, Ibarrola O, Mancilla S, Sarno F, Aguirre JJ, Razafindrakoto S, Southern P, Terán FJ, Keogh A, Salas G, Prina-Mello A, Lacal JC, Del Pozo A, Pankhurst QA, Hidalgo M, Gazeau F, Somoza Á, Schwartz S, Abasolo I. Preclinical Development of Magnetic Nanoparticles for Hyperthermia Treatment of Pancreatic Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:2924-2939. [PMID: 39745145 DOI: 10.1021/acsami.4c16129] [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: 01/18/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a very challenging disease with a very poor prognosis. It is characterized by a dense desmoplastic stroma that hampers drug penetration and limits the effectiveness of conventional chemotherapy (CT). As an alternative, the combination of CT with hyperthermia (HT) has been proposed as an innovative treatment modality for PDAC. In previous works, we reported on the development of iron oxide magnetic nanoparticles (MNPs) that, when exposed to time-varying magnetic fields, exhibit strong HT responses that inhibited the growth of pancreatic cancers. We report here on advances toward the clinical use of these MNPs as an intratumorally administered sterile magnetic fluid (the "NoCanTher ThermoTherapy" or "NTT" Agent) alongside intravenous standard-of-care drugs (gemcitabine and nab-paclitaxel) for the treatment of PDAC. In vitro cell viability assays show that the combination of low doses of CT and HT is highly synergistic, particularly in the BxPC-3 cell line. In vivo, biodistribution assays showed that the NTT Agent MNPs remained mainly within the tumor, concentrated around areas with a high stromal component. Moreover, the combined CT/HT treatment shows clear advantages over CT alone in terms of drug penetration and reduction of the tumor volume, suggesting a potential direct effect of HT in the disruption of the interstitial stroma to facilitate the access of the drugs to malignant cells. These studies have led to the approval and commencement of a clinical investigational study at the Vall d'Hebron University Hospital (Barcelona, Spain) of the NTT Agent alongside CT in patients with locally advanced PDAC.
Collapse
Affiliation(s)
- Zamira V Díaz-Riascos
- Clinical Biochemistry, Drug Delivery & Therapy (CB-DDT), Vall d'Hebron Institute of Research (VHIR), 08035 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08035 Barcelona, Spain
- Functional Validation & Preclinical Research (FVPR), Unit20 ICTS Nanbiosis, Vall d'Hebron Institute of Research (VHIR), 08035 Barcelona, Spain
| | - Monserrat Llaguno-Munive
- Clinical Biochemistry, Drug Delivery & Therapy (CB-DDT), Vall d'Hebron Institute of Research (VHIR), 08035 Barcelona, Spain
| | - Nuria Lafuente-Gómez
- IMDEA Nanociencia, Unidad Asociada al Centro Nacional de Biotecnología (CSIC) 28049 Madrid, Spain
| | - Yurena Luengo
- IMDEA Nanociencia, Unidad Asociada al Centro Nacional de Biotecnología (CSIC) 28049 Madrid, Spain
| | - Sarah Holmes
- Nanomedicine and Molecular Imaging group, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin (TCD), Dublin 8 Dublin, Ireland
| | - Jeanne Volatron
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS, Université Paris Diderot, Paris 75205, cedex, France
| | | | - Sandra Mancilla
- Clinical Biochemistry, Drug Delivery & Therapy (CB-DDT), Vall d'Hebron Institute of Research (VHIR), 08035 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08035 Barcelona, Spain
- Functional Validation & Preclinical Research (FVPR), Unit20 ICTS Nanbiosis, Vall d'Hebron Institute of Research (VHIR), 08035 Barcelona, Spain
| | - Francesca Sarno
- Grupo de Oncología Traslacional, Hospital Universitario de Fuenlabrada, 28942 Madrid, Spain
| | | | - Sarah Razafindrakoto
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS, Université Paris Diderot, Paris 75205, cedex, France
| | | | - Francisco J Terán
- IMDEA Nanociencia, Unidad Asociada al Centro Nacional de Biotecnología (CSIC) 28049 Madrid, Spain
- Unidad Asociada de Nanobiotecnología (CNB-CSIC e IMDEA Nanociencia), 28049 Madrid, Spain
- Unidad de Nanomateriales Avanzados, IMDEA Nanociencia (Unidad de I+D+I Asociada al Instituto de Ciencia de Materiales de Madrid, CSIC), 28049 Madrid, Spain
| | - Anna Keogh
- Department of Histopathology, St. James's Hospital and Trinity College Dublin, Cancer Molecular Diagnostics, Dublin 8 Dublin, Ireland
| | - Gorka Salas
- IMDEA Nanociencia, Unidad Asociada al Centro Nacional de Biotecnología (CSIC) 28049 Madrid, Spain
- Unidad Asociada de Nanobiotecnología (CNB-CSIC e IMDEA Nanociencia), 28049 Madrid, Spain
- Unidad de Nanomateriales Avanzados, IMDEA Nanociencia (Unidad de I+D+I Asociada al Instituto de Ciencia de Materiales de Madrid, CSIC), 28049 Madrid, Spain
| | - Adriele Prina-Mello
- Nanomedicine and Molecular Imaging group, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin (TCD), Dublin 8 Dublin, Ireland
- Trinity St. James's Cancer Institute, School of Medicine (TCD) and St. James's Hospital, Dublin 8 Dublin, Ireland
- Laboratory for Biological Characterisation of Advanced Materials (LBCAM), TTMI, School of Medicine, Trinity College Dublin, Dublin 8 Dublin, Ireland
| | - Juan Carlos Lacal
- Grupo de Oncología Traslacional, Hospital Universitario de Fuenlabrada, 28942 Madrid, Spain
- Instituto de Investigaciones Biomédicas (IIB), CSIC, 28029 Madrid, Spain
| | - Angel Del Pozo
- Nanomedicine and Molecular Imaging group, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin (TCD), Dublin 8 Dublin, Ireland
| | - Quentin A Pankhurst
- Resonant Circuits Limited, London W1S 4BS, U.K
- Healthcare Biomagnetics Laboratory, University College London, London W1S 4BS, U.K
| | - Manuel Hidalgo
- Grupo de Oncología Traslacional, Hospital Universitario de Fuenlabrada, 28942 Madrid, Spain
| | - Florence Gazeau
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS, Université Paris Diderot, Paris 75205, cedex, France
| | - Álvaro Somoza
- IMDEA Nanociencia, Unidad Asociada al Centro Nacional de Biotecnología (CSIC) 28049 Madrid, Spain
- Unidad Asociada de Nanobiotecnología (CNB-CSIC e IMDEA Nanociencia), 28049 Madrid, Spain
| | - Simó Schwartz
- Clinical Biochemistry, Drug Delivery & Therapy (CB-DDT), Vall d'Hebron Institute of Research (VHIR), 08035 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08035 Barcelona, Spain
- Servei de Bioquímica, Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Ibane Abasolo
- Clinical Biochemistry, Drug Delivery & Therapy (CB-DDT), Vall d'Hebron Institute of Research (VHIR), 08035 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08035 Barcelona, Spain
- Functional Validation & Preclinical Research (FVPR), Unit20 ICTS Nanbiosis, Vall d'Hebron Institute of Research (VHIR), 08035 Barcelona, Spain
- Servei de Bioquímica, Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
- Instituto de Química Avanzada de Cataluña (IQAC), CSIC, 08034 Barcelona, Spain
| |
Collapse
|
135
|
Jobu Y, Nishigawa M, Furihata K, Furihata M, Uchida K, Taniuchi K. Inhibitory effects of the combination of rapamycin with gemcitabine plus paclitaxel on the growth of pancreatic cancer tumors. Hum Cell 2025; 38:44. [PMID: 39794664 PMCID: PMC11723851 DOI: 10.1007/s13577-024-01165-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/16/2024] [Indexed: 01/13/2025]
Abstract
We previously examined the antitumor effects of short interfering RNA nanoparticles targeting mammalian target of rapamycin (mTOR) in an orthotopic pancreatic cancer mouse model. We herein report the inhibitory effects of the mTOR inhibitor rapamycin on tumor growth in a novel established mouse model of pancreatic cancer using human pancreatic cancer cell line-derived organoids. Gemcitabine, 5-fluorouracil, and gemcitabine plus nab-paclitaxel are clinically used to treat advanced pancreatic cancer. In vitro assays showed that rapamycin strongly inhibited cell invasion, while gemcitabine, 5-fluorouracil, and gemcitabine plus paclitaxel primarily inhibited cell proliferation with minimal effects on invasion. In vivo mouse experiments demonstrated that rapamycin exhibited superior antitumor activity to S-1 (a metabolically activated prodrug of 5-fluorouracil) and another mTOR inhibitor, everolimus, while its efficacy was similar to that of gemcitabine plus paclitaxel (which was used instead of nab-paclitaxel due to concerns about allergic reactions in mice to human albumin) in a mouse model of pancreatic cancer using human pancreatic cancer cell line-derived organoids. Furthermore, the combination of rapamycin with gemcitabine plus paclitaxel exerted synergistic inhibitory effects on the growth of pancreatic cancer tumors. Although the inhibition of tumor growth was significantly stronger in everolimus-treated mice than in control mice, there were no additive anti-growth effects when combined with gemcitabine plus paclitaxel. The present results suggest that the combination of rapamycin with gemcitabine plus paclitaxel achieved the greatest reduction in tumor volumes in the mouse xenograft model and, thus, has significant clinical promise.
Collapse
Affiliation(s)
- Yuri Jobu
- Department of Gastroenterology and Hepatology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Miki Nishigawa
- Department of Gastroenterology and Hepatology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Kaoru Furihata
- Department of Pathology, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
| | - Mutsuo Furihata
- Department of Pathology, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
| | - Kazushige Uchida
- Department of Gastroenterology and Hepatology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Keisuke Taniuchi
- Department of Gastroenterology and Hepatology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan.
| |
Collapse
|
136
|
Aleixo G, Voruganti T, Sedhom R. Three lessons from "The Fox and the Grapes" to inform precision oncology in the older adult. J Geriatr Oncol 2025:102186. [PMID: 39794225 DOI: 10.1016/j.jgo.2025.102186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Affiliation(s)
- Gabriel Aleixo
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Teja Voruganti
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ramy Sedhom
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA, USA; Penn Center for Cancer Care Innovation, Abramson Cancer Center, Penn Medicine, Philadelphia, PA, USA
| |
Collapse
|
137
|
Fanijavadi S, Thomassen M, Jensen LH. Targeting Triple NK Cell Suppression Mechanisms: A Comprehensive Review of Biomarkers in Pancreatic Cancer Therapy. Int J Mol Sci 2025; 26:515. [PMID: 39859231 PMCID: PMC11765000 DOI: 10.3390/ijms26020515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with poor outcomes due to frequent recurrence, metastasis, and resistance to treatment. A major contributor to this resistance is the tumor's ability to suppress natural killer (NK) cells, which are key players in the immune system's fight against cancer. In PDAC, the tumor microenvironment (TME) creates conditions that impair NK cell function, including reduced proliferation, weakened cytotoxicity, and limited tumor infiltration. This review examines how interactions between tumor-derived factors, NK cells, and the TME contribute to tumor progression and treatment resistance. To address these challenges, we propose a new "Triple NK Cell Biomarker Approach". This strategy focuses on identifying biomarkers from three critical areas: tumor characteristics, TME factors, and NK cell suppression mechanisms. This approach could guide personalized treatments to enhance NK cell activity. Additionally, we highlight the potential of combining NK cell-based therapies with conventional treatments and repurposed drugs to improve outcomes for PDAC patients. While progress has been made, more research is needed to better understand NK cell dysfunction and develop effective therapies to overcome these barriers.
Collapse
Affiliation(s)
- Sara Fanijavadi
- Cancer Polyclinic, Levanger Hospital, 7601 Levanger, Norway
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark;
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, 5000 Odense, Denmark;
- Department of Clinical Research, University of Southern Denmark, 5230 Odense, Denmark
| | - Lars Henrik Jensen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark;
- Department of Oncology, Institute of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
| |
Collapse
|
138
|
Shi CW, VanderMeer TJ, Pudusseri A. Metastatic pancreatic cancer now in remission: a case report and literature review. Discov Oncol 2025; 16:26. [PMID: 39786582 PMCID: PMC11717766 DOI: 10.1007/s12672-025-01756-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy with the majority of patients presenting at a late stage with unresectable or metastatic disease. Even with first line treatment, median survival is approximately 11 months in patients with advanced PDAC. This report details the unique case of a patient that presented with peritoneal metastases from an adenocarcinoma of the body of the pancreas, had a remarkable response to palliative chemotherapy and is alive without evidence of disease 12 months following cessation of all active treatment. The initial diagnosis was 4 years ago and extensive resection of the primary was completed 2 years ago. The patient was started on standard FOLFIRINOX chemotherapy regimen, completed 6 cycles, but stopped Oxaliplatin and Irinotecan due to neuropathy and fatigue, on November 5, 2020, and transitioned to 5-fluorouracil (5-FU) and leucovorin. There was radiographic response and a notable decrease in tumor marker CA 19-9. On July 12, 2022 he underwent a multivisceral resection that included a radical anterograde modular pancreatico-splenectomy, partial gastrectomy, and splenic flexure colectomy with primary anastomosis due to tumor involvement of the posterior stomach and splenic flexure. Surgical pathology noted a moderately differentiated, grade 2 tumor staged ypT2 N0 M0. He continued the same adjuvant regimen of 5-FU and leucovorin for approximately 9 months with no new or recurrent disease on imaging. His CA 19-9 decreased within normal range after surgery and has remained within the normal limits. He remains on active surveillance. Overall, barring clear availability for targeted therapies, a metastatic PDAC of the tail may be considered to have a better prognosis than previously considered. FOLFIRINOX is the ideal treatment if the patient has a high-performance status, and PRODIGE 35 recommends 8 minimum cycles. However, in our case, the patient only tolerated 6 cycles and was still highly responsive. Despite a stage IV diagnosis, the primary tumor was resected in order to mitigate the risk for mutation and progression. Although rare, greater hope for patients with PDAC of the tail with favorable tumor biology responsive to FOLFIRINOX may contribute to increased surgical resection rates and improve survival rates.
Collapse
|
139
|
Álvarez-Gallego R, Pazo-Cid R, López de San Vicente B, Macarulla T, Martinez E, Garicano F, Hernández I, Granja M, Ghanem I, Martinez J, Ribera P, Diaz R, Martin Valadés JI, Angeles MC, Cubillo A. Real-world effectiveness and safety of second- or third-line pegylated liposomal irinotecan plus 5-fluorouracil and folinic acid in pancreatic ductal adenocarcinoma in Spain. Ther Adv Med Oncol 2025; 17:17588359241309828. [PMID: 39781240 PMCID: PMC11707772 DOI: 10.1177/17588359241309828] [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: 04/14/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Treatment with pegylated nanoliposomal irinotecan (nal-IRI) plus 5-fluorouracil/leucovorin (folinic acid; 5-FU/LV) has demonstrated remarkable efficacy for metastatic pancreatic ductal adenocarcinoma (PDAC) in clinical trials. However, real-world data on the effectiveness of nal-IRI+5-FU/LV is heterogeneous and is lacking in Spain. To assess the effectiveness and safety of nal-IRI+5-FU/LV in real-life PDAC patients in Spain. A multicenter retrospective study was conducted. Patients aged ⩾18 years who had received at least one cycle of nal-IRI+5-FU/LV as second- or third-line therapy for PDAC were included. The primary endpoint was overall survival (OS) from nal-IRI+5-FU/LV treatment initiation and OS from the diagnosis of metastatic disease (metOS). Overall, 200 evaluable patients were included (⩾3 metastatic sites: 22%; liver/lung metastases: 71.5%/36.9%; and Eastern Cooperative Oncology Group 0-1: 87% at nal-IRI+5FU/LV treatment initiation). Patients received a median of four cycles of nal-IRI+5FU/LV for 2.8 months (range 1.4-7.2), and the treatment was received in the second line by 80% of the patients. The median OS was 7.2 months (6- and 12-month OS rates: 58.1% and 28.9%, respectively), with 27.2% of the patients achieving OS ⩾12 months. The median metOS was 17.5 months, with 30.2% of the patients experiencing metOS ⩾ 24 months. The median progression-free survival (PFS) was 3.7 months (6- and 12-month PFS rate: 37.6% and 15.3%, respectively). The disease control rate was 35.5%. The median CA 19-9 levels decreased by at least 50% in 28.2% of the cases during treatment. Overall, 36% of the patients experienced at least one grade 3-4 adverse event during treatment, the most common being diarrhea (42.6%) and asthenia (30.9%). This real-world study shows that treatment with nal-IRI+5-FU/LV for advanced or metastatic PDAC affords benefit in terms of survival, radiological and CA 19-9 response, and PFS comparable to that reported in the clinical trial setting with a manageable safety profile.
Collapse
Affiliation(s)
- Rafael Álvarez-Gallego
- Centro Integral Oncológico Clara Campal HM CIOCC, Hospital Universitario HM Sanchinarro, C\ Oña Nº10, Madrid 28050, Spain
- Facultad HM Hospitales de Ciencias de la Salud, Universidad Camilo José Cela (UCJC), Madrid, Spain
| | | | | | - Teresa Macarulla
- Vall d’Hebrón Institute of Oncology (VHIO), Hospital Universitario Vall d’Hebrón, Barcelona, Spain
| | - Eva Martinez
- Hospital Universitario Marques de Valdecilla, Santander, Spain
| | | | | | | | | | - Joaquina Martinez
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Paula Ribera
- Hospital Universitario Parc Tauli, Sabadell, Barcelona, Spain
| | - Roberto Diaz
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | | | - Antonio Cubillo
- Centro Integral Oncológico Clara Campal HM CIOCC, Hospital Universitario HM Sanchinarro, Madrid, Spain
- Facultad HM Hospitales de Ciencias de la Salud, Universidad Camilo José Cela (UCJC), Madrid, Spain
| |
Collapse
|
140
|
Carbone C, De Luca R, Puca E, Agostini A, Caggiano A, Priori L, Esposito A, Ascrizzi S, Piro G, Salvatore L, De Sanctis F, Ugel S, Corbo V, Neri D, Tortora G. Antibody-based delivery of interleukin-2 modulates the immunosuppressive tumor microenvironment and achieves cure in pancreatic ductal adenocarcinoma syngeneic mice. J Exp Clin Cancer Res 2025; 44:7. [PMID: 39773310 PMCID: PMC11705946 DOI: 10.1186/s13046-024-03238-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/24/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and deadly type of cancer, with an extremely low five-year overall survival rate. To date, current treatment options primarily involve various chemotherapies, which often prove ineffective and are associated with substantial toxicity. Furthermore, immunotherapies utilizing checkpoint inhibitors have shown limited efficacy in this context, highlighting an urgent need for novel therapeutic strategies. This study investigates the preclinical efficacy of an innovative targeted therapy based on antibody-cytokine fusion proteins, specifically interleukin-2 (IL-2), a pivotal driver of cell-mediated immunity, fused to L19 antibody, which selectively binds to extra domain B of fibronectin (EDB-FN1) expressed in the tumor microenvironment. METHODS We tested the effectiveness of different immunocytokines through in vivo characterization in syngeneic C57BL/6J orthotopic mouse models of PDAC. Based on these results, we decided to focus on L19-IL2. To assess the efficacy of this immunocytokine we developed an ex-vivo immune-spheroid interaction platform derived from murine 3D pancreatic cultures, and telomerase reverse transcriptase (TERT) specific T-lymphocytes. Moreover, we evaluated the anti-cancer effect of L19-IL2 in combination with standard therapy in vivo experiments in PDAC mouse models. Tumor samples collected after the treatments were characterized for tumor infiltrating immune cell components by bulk RNA sequencing (RNA-seq) and spatial transcriptomics (Stereo-seq) analysis. RESULTS The tumor-targeted L19-IL2 fusion protein demonstrated potent, dose-dependent anti-tumor activity in mice with pancreatic tumors resistant to standard chemotherapy. Spatial Transcriptomics (ST) and RNA-seq analyses indicated that L19-IL2 treatment induced a significant influx of immune cells into the tumor microenvironment, with these cells expressing activation markers like granzymes, perforins, and the IL-2 receptors. CONCLUSIONS Our results demonstrated that L19-IL2 enhances immune infiltration and cytotoxicity, remodeling the "cold" tumor microenvironment (TME) in PDAC. This innovative antibody-cytokine fusion protein improves therapeutic outcomes, paving the way for novel targeted treatment strategies in PDAC.
Collapse
Affiliation(s)
- Carmine Carbone
- Department of Medical and Surgical Sciences, Medical Oncology , Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Roberto De Luca
- Philochem AG, Libernstrasse 3, Otelfingen, 8112, Switzerland
| | - Emanuele Puca
- Philochem AG, Libernstrasse 3, Otelfingen, 8112, Switzerland
| | - Antonio Agostini
- Department of Medical and Surgical Sciences, Medical Oncology , Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Alessia Caggiano
- Department of Translational Medicine, Medical Oncology , Catholic University of the Sacred Heart, Rome, Italy
| | - Lorenzo Priori
- Department of Translational Medicine, Medical Oncology , Catholic University of the Sacred Heart, Rome, Italy
| | - Annachiara Esposito
- Department of Translational Medicine, Medical Oncology , Catholic University of the Sacred Heart, Rome, Italy
| | - Serena Ascrizzi
- Department of Translational Medicine, Medical Oncology , Catholic University of the Sacred Heart, Rome, Italy
| | - Geny Piro
- Department of Medical and Surgical Sciences, Medical Oncology , Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Lisa Salvatore
- Department of Medical and Surgical Sciences, Medical Oncology , Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
- Department of Translational Medicine, Medical Oncology , Catholic University of the Sacred Heart, Rome, Italy
| | - Francesco De Sanctis
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Stefano Ugel
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Vincenzo Corbo
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, CH-8093, Switzerland
- Philogen Spa, Piazza La Lizza 7, Siena, 53100, Italy
| | - Giampaolo Tortora
- Department of Medical and Surgical Sciences, Medical Oncology , Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy.
- Department of Translational Medicine, Medical Oncology , Catholic University of the Sacred Heart, Rome, Italy.
| |
Collapse
|
141
|
Takemura K, Ikeda K, Miyake H, Sogame Y, Yasuda H, Okada N, Iwata K, Sakagami J, Yamaguchi K, Itoh Y, Umemura A. Epithelial-Mesenchymal Transition Suppression by ML210 Enhances Gemcitabine Anti-Tumor Effects on PDAC Cells. Biomolecules 2025; 15:70. [PMID: 39858464 PMCID: PMC11763895 DOI: 10.3390/biom15010070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers in the world. Neoadjuvant chemotherapy (NAC) has become a standard treatment for patients scheduled for surgical resection, but the high rate of postoperative recurrence is a critical problem. Optimization of NAC is desirable to reduce postoperative recurrence and achieve long-term survival. However, if a patient's general condition deteriorates due to NAC toxicity, surgical outcomes may be compromised. Therefore, we aimed to identify drug(s) that can be used in combination with gemcitabine (GEM), a drug widely used for the treatment of PDAC, to inhibit distant metastatic recurrence, particularly after surgery. After several screening steps, ML210, a low molecular weight chemical, was found to suppress the epithelial-mesenchymal transition (EMT) in PDAC cells in combination with GEM. Specifically, low dose ML210 in combination with GEM was sufficient for cell migration without apparent toxicity or cell death. Mechanistically, ML210, which was developed as a glutathione peroxidase 4 (GPX4) inhibitor to induce lipid peroxidation, increased the oxidized lipid concentrations in PDAC cells. The oxidization of the cell membrane lipids may suppress EMT, including cell migration. Since EMT is a major malignant phenotype of PDAC, our findings may lead to the advancement of PDAC therapy, especially in the prevention of postoperative recurrence.
Collapse
Affiliation(s)
- Keisuke Takemura
- Department of Pharmacology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (K.T.); (K.I.); (N.O.); (K.I.)
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
| | - Kyohei Ikeda
- Department of Pharmacology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (K.T.); (K.I.); (N.O.); (K.I.)
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
| | - Hayato Miyake
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
| | - Yoshio Sogame
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
| | - Hiroaki Yasuda
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
- Saiseikai Shiga Hospital, 2-4-1 Ohashi, Ritto 520-3046, Shiga, Japan
| | - Nobuhiro Okada
- Department of Pharmacology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (K.T.); (K.I.); (N.O.); (K.I.)
| | - Kazumi Iwata
- Department of Pharmacology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (K.T.); (K.I.); (N.O.); (K.I.)
| | - Junichi Sakagami
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
- Fukuchiyama City Hospital, 231 Atsunaka-cho, Fukuchiyama 620-8505, Kyoto, Japan
| | - Kanji Yamaguchi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
| | - Atsushi Umemura
- Department of Pharmacology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (K.T.); (K.I.); (N.O.); (K.I.)
| |
Collapse
|
142
|
Wnuk J, Hudy D, Strzelczyk JK, Michalecki Ł, Dybek K, Gisterek-Grocholska I. Serum hsa-miR-22-3p, hsa-miR-885-5p, Lipase-to-Amylase Ratio, C-Reactive Protein, CA19-9, and Neutrophil-to-Lymphocyte Ratio as Prognostic Factors in Advanced Pancreatic Ductal Adenocarcinoma. Curr Issues Mol Biol 2025; 47:27. [PMID: 39852142 PMCID: PMC11763715 DOI: 10.3390/cimb47010027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/28/2024] [Accepted: 01/01/2025] [Indexed: 01/26/2025] Open
Abstract
Pancreatic cancer (PC) is the seventh most common cause of cancer-related death worldwide. The low survival rate may be due to late diagnosis and asymptomatic early-stage disease. Most patients are diagnosed at an advanced stage of the disease. The search for novel prognostic factors is still needed. Two miRNAs, miR-22-3p and miR-885-5p, which show increased expression in PC, were selected for this study. The aim of this study was to evaluate the utility of these miRNAs in the prognosis of PC. Other prognostic factors such as lipase-to-amylase ratio (LAR), neutrophil-to-lymphocyte ratio (NLR), and carbohydrate antigen 19-9 (CA19-9) were also evaluated in this study. This study was conducted in 50 patients previously diagnosed with pancreatic ductal adenocarcinoma in clinical stage (CS) III and IV. All patients underwent a complete medical history, physical examination, and routine laboratory tests including a complete blood count, C-reactive protein (CRP), CA19-9, lipase, and amylase. Two additional blood samples were taken from each patient to separate plasma and serum. Isolation of miRNA was performed using TRI reagent with cel-miR-39-3p as a spike-in control. Reverse transcription of miRNA was performed using a TaqMan Advanced miRNA cDNA Synthesis Kit. The relative expression levels of miR-22-3p and miR-885-5p were measured using RT-qPCR. Serum hsa-miR-22-3p was detected in 22 cases (44%), while hsa-miR-885-5p was detected in 33 cases (66%). There were no statistically significant differences in serum or plasma miRNA expression levels between patient groups based on clinical stage, gender, or BMI. There were no statistically significant differences in LAR between patients with different CS. For NLR, CRP and CA19-9 thresholds were determined using ROC analysis (6.63, 24.7 mg/L and 4691 U/mL, respectively). Cox's F test for overall survival showed statistically significant differences between groups (p = 0.002 for NLR, p = 0.007 for CRP and p = 0.007 for CA19-9). Utility as prognostic biomarkers was confirmed in univariate and multivariate analysis for CA19-9, CRP, and NLR. The selected miRNAs and LAR were not confirmed as reliable prognostic markers in PC.
Collapse
Affiliation(s)
- Jakub Wnuk
- Department of Oncology and Radiotherapy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 35 Ceglana St., 40-515 Katowice, Poland; (J.W.)
| | - Dorota Hudy
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland; (D.H.); (J.K.S.)
| | - Joanna Katarzyna Strzelczyk
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland; (D.H.); (J.K.S.)
| | - Łukasz Michalecki
- Department of Oncology and Radiotherapy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 35 Ceglana St., 40-515 Katowice, Poland; (J.W.)
| | - Kamil Dybek
- Central Laboratory, University Clinical Center, Medical University of Silesia in Katowice, 14 Medyków St., 40-752 Katowice, Poland
| | - Iwona Gisterek-Grocholska
- Department of Oncology and Radiotherapy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 35 Ceglana St., 40-515 Katowice, Poland; (J.W.)
| |
Collapse
|
143
|
Aseafan M, Alfakeeh AH, Tashkandi E, Mahrous M, Alghamdi M, Alshamsan B, Al-Hajeili M, Bakhsh S, Alshammari K, Almugbel FA, Alfagih AH, Allehebi A, Montaser M, Elsafty MH, Elnaghi KAE, Issa I, Bakshi E, AlSubaie S, AlMutairi B, Mokhtar H, Aboelatta M, Bukhari N, Alzahrani AM, Elhassan T, Alqahtani A, Bazarbashi S. Real-world clinical outcome of unresectable locally advanced & de-novo metastatic pancreatic ductal adenocarcinoma: a multicentre retrospective study. BMC Cancer 2025; 25:7. [PMID: 39754118 PMCID: PMC11697791 DOI: 10.1186/s12885-024-13386-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with limited treatment options yielding poor outcomes. This study aimed to evaluate the real-world clinical characteristics, treatment patterns, and outcomes of patients with locally advanced unresectable and de-novo metastatic PDAC in Saudi Arabia, providing regional data to compare with international benchmarks. METHODS This is a retrospective, multicentre study involving 350 patients diagnosed with unresectable locally advanced or de-novo metastatic PDAC between January 2015 and November 2023. Data were collected from 10 oncology centers across Saudi Arabia. RESULTS The median age at diagnosis was 60 years, with 63% of patients presenting with multiple metastatic sites, primarily in the liver (66.3%). FOLFIRINOX was the most common first-line treatment (55.1%), followed by gemcitabine plus nab-paclitaxel (15.1%). The median PFS for first-line treatment was 5.3 months, with FOLFIRINOX achieving the longest PFS (6.5 months). The median OS was 10.34 months for the entire cohort, with better survival outcomes observed in patients receiving FOLFIRINOX (12.3 months). Independent prognostic factors for PFS and OS included performance status, first-line regimen, and neutrophil-lymphocyte ratio (NLR). Among patients tested, 7.1% had deficient mismatch repair (d-MMR), and 5.8% harbored BRCA mutations. CONCLUSIONS This real-world study confirms that clinical outcomes for locally advanced unresectable and metastatic PDAC in Saudi Arabia are consistent with international data, with FOLFIRINOX showing superior outcomes over gemcitabine-based regimens. However, both treatments reflect the persistent poor prognosis of PDAC, underscoring the need for novel therapeutic strategies. Further research is warranted to optimize treatment selection and improve survival outcomes in this population.
Collapse
Affiliation(s)
- Mohamed Aseafan
- Section of Medical Oncology, Department of Internal Medicine, Security Forces Hospital, Riyadh, Saudi Arabia.
| | - Ali H Alfakeeh
- Comprehensive Cancer Center, Medical Oncology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Emad Tashkandi
- College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mervat Mahrous
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
- Collage of Medicine, Minia University, Minia, Egypt
| | - Mohammed Alghamdi
- Oncology Center, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Bader Alshamsan
- Department of Medicine, College of Medicine, Qassim University, Buraydah, Saudi Arabia
- Prince Faisal Cancer Center, King Fahad Specialist Hospital, Qassim Health Clusster, Buraydah, Saudi Arabia
| | - Marwan Al-Hajeili
- Department of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Safwan Bakhsh
- Department of Medical Oncology, King Faisal Specialist Hospital and Research Center-Jeddah, Jeddah, Saudi Arabia
| | - Kanan Alshammari
- Department of Medical Oncology, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Fahad A Almugbel
- Department of Medical Oncology, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Abdulhameed H Alfagih
- Comprehensive Cancer Center, Medical Oncology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ahmed Allehebi
- Department of Medical Oncology, King Faisal Specialist Hospital and Research Center-Jeddah, Jeddah, Saudi Arabia
| | - Mohamed Montaser
- Section of Medical Oncology, Department of Internal Medicine, Security Forces Hospital, Riyadh, Saudi Arabia
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | - Ibrahim Issa
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Eesa Bakshi
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Sadeem AlSubaie
- Pathology and Laboratory Medicine, Security Forces Hospital, Riyadh, Saudi Arabia
| | - Bandar AlMutairi
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Hoda Mokhtar
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Mohamed Aboelatta
- Prince Faisal Cancer Center, King Fahad Specialist Hospital, Qassim Health Clusster, Buraydah, Saudi Arabia
| | - Nedal Bukhari
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Ali M Alzahrani
- Comprehensive Cancer Center, Medical Oncology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Tusneem Elhassan
- Research Unit, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ali Alqahtani
- Department of Medical Oncology, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Shouki Bazarbashi
- Department of Medical Oncology, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
144
|
Liu X, Wang X, Ren J, Fang Y, Gu M, Zhou F, Xiao R, Luo X, Bai J, Jiang D, Tang Y, Ren B, You L, Zhao Y. Machine learning based identification of an amino acid metabolism related signature for predicting prognosis and immune microenvironment in pancreatic cancer. BMC Cancer 2025; 25:6. [PMID: 39754071 PMCID: PMC11697724 DOI: 10.1186/s12885-024-13374-4] [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: 06/10/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Pancreatic cancer is a highly aggressive neoplasm characterized by poor diagnosis. Amino acids play a prominent role in the occurrence and progression of pancreatic cancer as essential building blocks for protein synthesis and key regulators of cellular metabolism. Understanding the interplay between pancreatic cancer and amino acid metabolism offers potential avenues for improving patient clinical outcomes. METHODS A comprehensive analysis integrating 10 machine learning algorithms was executed to pinpoint amino acid metabolic signature. The signature was validated across both internal and external cohorts. Subsequent GSEA was employed to unveil the enriched gene sets and signaling pathways within high- and low-risk subgroups. TMB and drug sensitivity analyses were carried out via Maftools and oncoPredict R packages. CIBERSORT and ssGSEA were harnessed to delve into the immune landscape disparities. Single-cell transcriptomics, qPCR, and Immunohistochemistry were performed to corroborate the expression levels and prognostic significance of this signature. RESULTS A four gene based amino acid metabolic signature with superior prognostic capabilities was identified by the combination of 10 machine learning methods. It showed that the novel prognostic model could effectively distinguish patients into high- and low-risk groups in both internal and external cohorts. Notably, the risk score from this novel signature showed significant correlations with TMB, drug resistance, as well as a heightened likelihood of immune evasion and suboptimal responses to immunotherapeutic interventions. CONCLUSION Our findings suggested that amino acid metabolism-related signature was closely related to the development, prognosis and immune microenvironment of pancreatic cancer.
Collapse
Affiliation(s)
- Xiaohong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Xing Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Jie Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Yuan Fang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Minzhi Gu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Feihan Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Ruiling Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Xiyuan Luo
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Jialu Bai
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Decheng Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Yuemeng Tang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Bo Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking, Beijing, 100023, People's Republic of China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China.
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China.
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking, Beijing, 100023, People's Republic of China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China.
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking, Beijing, 100023, People's Republic of China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China.
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China.
| |
Collapse
|
145
|
Norton C, Shaw MS, Rubnitz Z, Smith J, Soares HP, Nevala-Plagemann CD, Garrido-Laguna I, Florou V. KRAS Mutation Status and Treatment Outcomes in Patients With Metastatic Pancreatic Adenocarcinoma. JAMA Netw Open 2025; 8:e2453588. [PMID: 39777438 PMCID: PMC11707629 DOI: 10.1001/jamanetworkopen.2024.53588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/05/2024] [Indexed: 01/11/2025] Open
Abstract
Importance Despite the high prevalence of KRAS alterations in pancreatic ductal adenocarcinoma (PDAC), the clinical impact of common KRAS mutations with different cytotoxic regimens is unknown. This evidence is important to inform current treatment and provide a benchmark for emergent targeted KRAS therapies in metastatic PDAC. Objective To assess the clinical implications of common KRAS G12 mutations in PDAC and to compare outcomes of standard-of-care multiagent therapies across these common mutations. Design, Setting, and Participants This retrospective cohort study obtained deidentified clinical data for 5382 patients from a nationwide (US-based) clinicogenomic database. The deidentified data originated from approximately 280 US cancer clinics (approximately 800 sites of care). Patients diagnosed with metastatic PDAC from February 9, 2010, to September 20, 2022, and with sufficient follow-up and treatment data were included. Main Outcomes and Measures Median overall survival (OS) and time to next treatment (TTNT) were calculated for each KRAS mutation group. Hazard ratios (HRs) were generated using multivariate Cox proportional hazards models for KRAS mutations and mutation-treatment combinations. Results A total of 2433 patients with PDAC were included in the analysis (mean age at first treatment, 67.0 [range, 66.0-68.0] years; 1340 male [55.1%]). Among 2023 patients with KRAS mutations, those with G12R had the longest median TTNT (6.0 [95% CI, 5.2-6.6] months) and the longest median OS (13.2 [95% CI, 10.6-15.2] months). Patients with KRAS G12D and G12V mutations had a significantly higher risk of disease progression (HRs, 1.15; [95% CI, 1.04-1.29; P = .009] and 1.16 [95% CI, 1.04-1.30; P = .01], respectively) and death (HRs, 1.29 [95% CI, 1.15-1.45; P < .001] and 1.23 [95% CI, 1.09-1.39; P < .001], respectively) compared with KRAS wild type. The FOLFIRINOX regimen (fluorouracil, irinotecan, oxaliplatin, and leucovorin) had a significantly lower risk of treatment progression and death than gemcitabine with (HRs, 1.19 [95% CI, [1.09-1.29; P < .001] and 1.18 [95% CI, 1.07-1.29; P < .001], respectively) or without (HRs, 1.37 [95% CI, 1.11-1.69; P = .003] and 1.41 [95% CI 1.13-1.75; P = .002], respectively) nab-paclitaxel across all patients. Conclusions and Relevance In this cohort study of 2433 patients with PDAC, KRAS G12D and G12V mutations were associated with worse patient outcomes compared with KRAS wild type. KRAS G12R was associated with more favorable patient outcomes, and FOLFIRINOX was associated with better patient outcomes than gemcitabine-based therapies. These findings highlight the need for more effective systemic therapies for these groups of patients.
Collapse
Affiliation(s)
- Carter Norton
- Huntsman Cancer Institute, University of Utah Health Care, Salt Lake City
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Zachary Rubnitz
- Department of Internal Medicine, Division of Oncology, University of Utah Health Care, Salt Lake City
- Department of Internal Medicine, University of Utah Health Care, Salt Lake City
| | - Jarrod Smith
- Department of Internal Medicine, Division of Oncology, University of Utah Health Care, Salt Lake City
- Department of Internal Medicine, University of Utah Health Care, Salt Lake City
| | - Heloisa P. Soares
- Huntsman Cancer Institute, University of Utah Health Care, Salt Lake City
- Department of Internal Medicine, Division of Oncology, University of Utah Health Care, Salt Lake City
| | - Christopher D. Nevala-Plagemann
- Huntsman Cancer Institute, University of Utah Health Care, Salt Lake City
- Department of Internal Medicine, Division of Oncology, University of Utah Health Care, Salt Lake City
| | - Ignacio Garrido-Laguna
- Huntsman Cancer Institute, University of Utah Health Care, Salt Lake City
- Department of Internal Medicine, Division of Oncology, University of Utah Health Care, Salt Lake City
| | - Vaia Florou
- Huntsman Cancer Institute, University of Utah Health Care, Salt Lake City
- Department of Internal Medicine, Division of Oncology, University of Utah Health Care, Salt Lake City
| |
Collapse
|
146
|
Shinohara A, Takumoto Y, Tauchi J, Morishita K, Kawasaki T, Akazawa M. Cost-effectiveness of systematic chemotherapy for metastatic pancreatic cancer: a retrospective study using Japanese clinical data. Sci Rep 2025; 15:267. [PMID: 39747263 PMCID: PMC11696500 DOI: 10.1038/s41598-024-80549-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 11/19/2024] [Indexed: 01/04/2025] Open
Abstract
We compared the cost-effectiveness of gemcitabine plus nab-paclitaxel (GnP) and modified FOLFIRINOX (mFFX)-standard first-line treatments for metastatic pancreatic cancer in Japan. This retrospective cohort study included patients with metastatic pancreatic cancer treated at the National Cancer Center Hospital East in Japan between December 2013 and February 2017. A partitioned survival model, featuring five mutually exclusive health states, was developed. A cost-effectiveness analysis was performed to obtain total costs and quality-adjusted life-years (QALYs). A 2% annual discount rate was applied to costs and outcomes. Parameter uncertainty effect was assessed using a one-way sensitivity analysis. mFFX treatment involved intravenous infusions of oxaliplatin, levofolinate, and irinotecan, whereas GnP treatment involved weekly intravenous administration of nab-paclitaxel and gemcitabine. Dosages were adjusted based on observed adverse events. The total costs of the mFFX and GnP arms were JPY 3,193,597 (USD 31,006) and JPY 3,913,171 (USD 37,992), respectively. Incremental total costs of JPY 719,574 (USD 6,986) were associated with GnP compared to mFFX. Total QALYs were 0.427 and 0.435 for mFFX and GnP, respectively. Therefore, GnP has slightly higher QALYs than mFFX in Japanese real-world clinical practice, suggesting it has a potentially better safety profile, although the impact on healthcare costs persists.
Collapse
Affiliation(s)
- Akira Shinohara
- Department of Pharmacy, National Cancer Center Hospital East, Chiba, Japan
| | - Yuki Takumoto
- Department of Public Health and Epidemiology, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Junko Tauchi
- Department of Pharmacy, National Cancer Center Hospital East, Chiba, Japan
| | - Koki Morishita
- Department of Pharmacy, National Cancer Center Hospital East, Chiba, Japan
| | | | - Manabu Akazawa
- Department of Public Health and Epidemiology, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan.
| |
Collapse
|
147
|
Lee W, Song G, Bae H. In vitro and in silico study of the synergistic anticancer effect of alpinumisoflavone with gemcitabine on pancreatic ductal adenocarcinoma through suppression of ribonucleotide reductase subunit-M1. Eur J Pharm Sci 2025; 204:106969. [PMID: 39577749 DOI: 10.1016/j.ejps.2024.106969] [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: 07/12/2024] [Revised: 10/21/2024] [Accepted: 11/20/2024] [Indexed: 11/24/2024]
Abstract
A highly aggressive neoplastic disease, pancreatic ductal adenocarcinoma (PDAC) is documented as the third chief cause of cancer-associated mortality in both sexes combined in the United States. For decades, gemcitabine-based chemotherapy has been embraced as a cornerstone drug for the treatment of PDAC. However, there have been several unsolved problems, including cytotoxicity, and chemoresistance. Gemcitabine efficacy was attributed to the attenuation of ribonucleotide reductase subunit-M1 (RRM1). Overexpression of RRM1 in PDAC is highly correlated with gemcitabine resistance and reduced gemcitabine sensitivity, resulting in a poor survival rate even after gemcitabine treatment. Moreover, the status of TP53, a tumor suppressor gene, assumes a decisive role in the response of PDAC to gemcitabine. Therefore, targeting RRM1 and P53 might be a therapeutic strategy for strengthening gemcitabine efficacy and cytotoxicity against PDAC. Alpinumisoflavone (AIF) is a prenylated isoflavone originated in Cudrania tricuspidate with versatile bioactive properties, including anticancer activity. However, there was no report whether AIF can exert anticancer effect and exhibit synergistic effect with gemcitabine against PDAC. Therefore, the anticancer properties of AIF were assessed with PANC-1 and MIA PaCa-2. In addition, synergism between AIF and gemcitabine were analyzed. Moreover, the contribution of P53 and RRM1 expression to gemcitabine resistance was assessed by comparing their protein levels in PDAC cells and normal pancreatic cells. The interactions of AIF with RRM1 protein were confirmed by molecular docking and dynamics simulation. Therefore, AIF enhances gemcitabine efficacy against PDAC through the regulation of P53 and RRM1.
Collapse
Affiliation(s)
- Woonghee Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, South Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, South Korea.
| | - Hyocheol Bae
- Department of Oriental Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, South Korea.
| |
Collapse
|
148
|
Jogi M, Asnani H, Singh S, Kumar P. Nimbolide: A Potential Phytochemical Agent in Multimodal Pancreatic Cancer Therapies. Mini Rev Med Chem 2025; 25:27-41. [PMID: 38874049 DOI: 10.2174/0113895575293138240527061556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/09/2024] [Accepted: 04/20/2024] [Indexed: 06/15/2024]
Abstract
A significant contributor to cancer-related death, pancreatic cancer (PC) has a terrible prognosis in general that has not altered over many years. Currently, it is extremely difficult to prevent disease or discover it early enough to initiate treatment. PC is a challenging malignancy to treat, and several major impediments significantly impact the effectiveness of its treatment. These obstacles primarily include chemoresistance, drug toxicity, and limited drug bioavailability. Phytochemicals can be used as an alternative to chemotherapeutic drugs, or they can augment the anticancer properties of the chemotherapeutic agents. Nimbolide (NL) is a prominent limonoid compound found in Azadirachta indica, and has garnered substantial attention as a phytochemical with anticancer potential. It has powerful antiproliferative effects on a variety of cancer cell lines and is effective as a chemotherapeutic in preclinical studies. The primary modes of action of NL include suppression of metastasis and angiogenesis, activation of apoptosis, anti-proliferation, and control of enzymes that metabolize carcinogens. Despite numerous pharmacodynamic (PD) investigations, NL is still in the early stages of the drug development process because no comprehensive pharmacokinetic studies or long-term toxicity studies. Preclinical and toxicological assessments should be conducted to establish an appropriate dosage range, ensuring the safety of NL for its application in initial human clinical trials. This review endeavors to provide a comprehensive summary of the current developmental stage of NL along with nanoparticles as a principal candidate for therapeutic purposes in PC.
Collapse
Affiliation(s)
- Mukesh Jogi
- Division of Molecular Biology, ICMR-National Institute of Cancer Prevention and Research (NICPR), NOIDA, India
- Amity Institute of Biotechnology, Amity University, NOIDA, India
- Department of Biotecnology ICMR- National Institute for Research in Environmental Health, Bhopal, India
| | - Hitakshi Asnani
- Banasthli Vidyapith, Radha Kishnpura, Rajasthan, 304022, India
| | - Sohini Singh
- Amity Institute of Biotechnology, Amity University, NOIDA, India
| | - Pramod Kumar
- Division of Molecular Biology, ICMR-National Institute of Cancer Prevention and Research (NICPR), NOIDA, India
| |
Collapse
|
149
|
Han S, Tushoski-Alemán GW, Zhang P, Zheng G, Zhou D, Huo Z, Licht J, George TJ, Allegra C, Trevino JG, Hughes SJ. A novel regimen for pancreatic ductal adenocarcinoma targeting MEK, BCL-xL, and EGFR. Neoplasia 2025; 59:101070. [PMID: 39541736 PMCID: PMC11609319 DOI: 10.1016/j.neo.2024.101070] [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: 09/11/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
Oncogenic KRAS signaling plays a critical role in pancreatic ductal adenocarcinoma (PDAC) biology. Recent studies indicate that the combination of MEK and BCL-xL inhibition is synthetically lethal and holds promise for some types of solid cancers, however, patient response was poorly observed in PDAC predominantly due to amplified EGFR signaling. Here, we leverage the advantage of the combinational treatment strategy and designed a triplet regimen targeting the comprehensive RAS activation networks through simultaneously blocking MEK/BCL-xL/EGFR. The cytotoxicity of trametinib (MEK inhibitor), DT2216 (BCL-xL degrader) and afatinib (pan-EGFR inhibitor) and their combination was tested in patient-derived, primary PDAC cells using a live cell imaging system. Patient-derived xenograft (PDX) model was employed for the evaluation of the therapeutic efficacy and safety of the combinational regimen. Targeted pathway cascades activities were analyzed using multiplex phosphor-immune assays. In vitro comparisons showed the addition of afatinib as a third agent was statistically superior compared to a doublet of trametinib+DT2216 in suppressing cell growth and inducing cell death in all cell lines tested. This triplet similarly demonstrated significant superiority over the doublet of MEK/BCL-xL inhibition in the in vivo murine model. The triplet regimen was well tolerated in vivo. Overall tumor growth rates were significantly reduced in doublet treatment compared to controls, and further reduced in the triplet treatment group. Pathway analysis revealed the addition of afatinib in triplet regimen further inhibited PI3K/AKT effectors of p90RSK, p70S6K, and GSK3α/β along with a secondary pathway of P38 MAPK. Our study identifies an important contribution of EGFR inhibition to elevate the response of PDAC, supporting a clinical assessment of this triplet combination in patients.
Collapse
Affiliation(s)
- Song Han
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Gerik W Tushoski-Alemán
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Peiyi Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Daohong Zhou
- Department of Biochemistry and Structural Biology & Center for Innovative Drug Discovery, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Zhiguang Huo
- Department of Biostatistics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jonathan Licht
- UF Health Cancer Center, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Thomas J George
- UF Health Cancer Center, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Carmen Allegra
- UF Health Cancer Center, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jose G Trevino
- Division of Surgical Oncology, Department of Surgery, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Steven J Hughes
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
150
|
Heckler M, Polychronidis G, Kinny-Köster B, Roth S, Hank T, Kaiser J, Michalski C, Loos M. Thrombosis and anticoagulation after portal vein reconstruction during pancreatic surgery: a systematic review. J Gastrointest Surg 2025; 29:101852. [PMID: 39406295 DOI: 10.1016/j.gassur.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/20/2024] [Accepted: 10/05/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Portal vein (PV) resection and reconstruction, which includes the resection and reconstruction of the PV and superior mesenteric vein, enable surgical removal of borderline resectable and locally advanced pancreatic cancer. Thrombosis of the reconstructed PV represents a major cause of early postoperative and long-term morbidity and mortality. No universally accepted standard for anticoagulation exists. This study aimed to assess early and late thrombosis rates after PV reconstruction with special regard to the type of PV reconstruction and anticoagulation regimen and to comprehensively assess thrombotic events and their clinical effect in patients receiving pancreatic surgery with venous resection and reconstruction. METHODS The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed. Studies reporting on PV resection and reconstruction providing data on thrombosis rates were included. The following parameters were assessed: study type, year of publication, number of patients, type/number of PV reconstruction, follow-up period, postoperative mortality, thrombosis rate of the reconstructed PV axis, intraoperative blood loss, and anticoagulation. RESULTS A total of 23 studies with 2751 patients were included in the final analysis. Of note, 670 patients received tangential resection of the PV with venorrhaphy or patch repair, 1505 patients had segmental resection with end-to-end reconstruction, and 576 patients received reconstruction with an interposition graft/conduit. The pooled overall thrombosis rate was 15%. Reconstruction of tangential defects with either venorrhaphy or patch repair and end-to-end repair of segmental defects resulted in a thrombosis rate of 12%. Subgroup analysis according to the type of graft reconstruction revealed the highest occlusion rates of 55% in patients with allogeneic grafts, followed by up to 27% in patients with synthetic PV conduits. Autologous conduits had a thrombosis rate of 10%. Early thrombotic events were detected in 5% of patients after venorrhaphy/patch reconstruction and end-to-end reconstruction. Early events were most common in the allogeneic graft subgroup (22%), followed by synthetic conduits (15%). There were fewer early events in the autologous graft group (7%). Early PV thrombosis was associated with relevant mortality of up to 26%. Anticoagulation regimens varied between studies. CONCLUSION The overall thrombosis rate after PV resection is low. However, among the different reconstruction techniques, allogeneic interposition grafts/conduits had the highest thrombosis rates among the different types of reconstruction after PV resection. No specific anticoagulation strategy can be considered beneficial based on the existing literature.
Collapse
Affiliation(s)
- Max Heckler
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Georgios Polychronidis
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany; Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Benedict Kinny-Köster
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Susanne Roth
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Hank
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Joerg Kaiser
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Christoph Michalski
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Loos
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|