1
|
Guenther M, Surendran SA, Steinke LM, Liou I, Palm MA, Heinemann V, Haas M, Boeck S, Ormanns S. The Prognostic, Predictive and Clinicopathological Implications of KRT81/HNF1A- and GATA6-Based Transcriptional Subtyping in Pancreatic Cancer. Biomolecules 2025; 15:426. [PMID: 40149962 PMCID: PMC11940166 DOI: 10.3390/biom15030426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Transcriptional subtypes of pancreatic ductal adenocarcinoma (PDAC) based on the expression of hallmark genes may have prognostic implications and potential predictive functions. The two most employed subtyping markers assess the combined expression of KRT81 and HNF1A or of GATA6 alone, which can be detected by immunohistochemistry (IHC). This study aimed to determine the prognostic or predictive impact of both subtyping marker panels in two large cohorts of advanced and resected pancreatic ductal adenocarcinoma (PDAC) patients. METHODS Transcriptional subtypes were determined by combining the expression of KRT81/HNF1A or assessing GATA6 expression alone by IHC in samples of two independent PDAC patient cohorts (advanced PDAC n = 139 and resected PDAC n = 411) as well as in 57 matched primary tumors and their corresponding metastases. RNAseq-based expression data of 316 resected PDAC patients was analyzed for validation. RESULTS Transcriptional subtypes widely overlapped in both marker panels (χ2p < 0.001) but switched during disease progression in up to 31.6% of patients. They had a modest impact on the patients' prognosis in both cohorts, with longer overall survival (OS) for patients with KRT81-/HNF1A+ or GATA6+ tumors but better progression-free survival (PFS) and disease-free survival (DFS) in patients with KRT81+/GATA6- tumors treated with palliative or adjuvant gemcitabine-based chemotherapy. RNAseq expression data confirmed the findings. CONCLUSIONS Transcriptional subtypes have differential responses to palliative and adjuvant gemcitabine-based chemotherapy and may change during disease progression. Both employed subtyping marker panels are equivalent and may be used to inform clinical therapy decisions.
Collapse
MESH Headings
- Humans
- GATA6 Transcription Factor/genetics
- GATA6 Transcription Factor/metabolism
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/mortality
- Female
- Male
- Prognosis
- Middle Aged
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Aged
- Hepatocyte Nuclear Factor 1-alpha/genetics
- Hepatocyte Nuclear Factor 1-alpha/metabolism
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/metabolism
- Gene Expression Regulation, Neoplastic
- Keratins, Hair-Specific/genetics
- Keratins, Hair-Specific/metabolism
- Keratins, Type II/genetics
- Keratins, Type II/metabolism
Collapse
Affiliation(s)
- Michael Guenther
- Innpath Institute of Pathology, Tirol Kliniken, 6020 Innsbruck, Austria;
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
| | - Sai Agash Surendran
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
| | - Lea Margareta Steinke
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
| | - Iduna Liou
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
| | - Melanie Alexandra Palm
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
| | - Volker Heinemann
- Department of Hematology and Oncology, München Klinik Neuperlach, 81737 Munich, Germany; (V.H.); (M.H.); (S.B.)
| | - Michael Haas
- Department of Hematology and Oncology, München Klinik Neuperlach, 81737 Munich, Germany; (V.H.); (M.H.); (S.B.)
- Department of Internal Medicine III, Grosshadern University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Stefan Boeck
- Department of Hematology and Oncology, München Klinik Neuperlach, 81737 Munich, Germany; (V.H.); (M.H.); (S.B.)
- Department of Internal Medicine III, Grosshadern University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Steffen Ormanns
- Innpath Institute of Pathology, Tirol Kliniken, 6020 Innsbruck, Austria;
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
2
|
Chen Y, Liang S, Chen B, Jiao F, Deng X, Liu X. Novel HSA-PMEMA Nanomicelles Prepared via Site-Specific In Situ Polymerization-Induced Self-Assembly for Improved Intracellular Delivery of Paclitaxel. Pharmaceutics 2025; 17:316. [PMID: 40142980 PMCID: PMC11945012 DOI: 10.3390/pharmaceutics17030316] [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: 01/03/2025] [Revised: 02/14/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Paclitaxel (PTX) is a potent anticancer drug that is poorly soluble in water. To enhance its delivery efficiency in aqueous environments, amphiphilic polymer micelles are often used as nanocarriers for PTX in clinical settings. However, the hydrophilic polymer segments on the surface of these micelles may possess potential immunogenicity, posing risks in clinical applications. To address this issue, nanomicelles based on human serum albumin (HSA)-hydrophobic polymer conjugates constructed via site-specific in situ polymerization-induced self-assembly (SI-PISA) are considered a promising alternative. The HSA shell not only ensures good biocompatibility but also enhances cellular uptake because of endogenous albumin trafficking pathways. Moreover, compared to traditional methods of creating protein-hydrophobic polymer conjugates, SI-PISA demonstrates higher reaction efficiency and better preservation of protein functionality. Methods: We synthesized HSA-PMEMA nanomicelles via SI-PISA using HSA and methoxyethyl methacrylate (MEMA)-a novel hydrophobic monomer with a well-defined and stable chemical structure. The protein activity and the PTX intracellular delivery efficiency of HSA-PMEMA nanomicelles were evaluated. Results: The CD spectra of HSA and HSA-PMEMA exhibited similar shapes, and the relative esterase-like activity of HSA-PMEMA was 94% that of unmodified HSA. Flow cytometry results showed that Cy7 fluorescence intensity in cells treated with HSA-PMEMA-Cy7 was approximately 1.35 times that in cells treated with HSA-Cy7; meanwhile, HPLC results indicated that, under the same conditions, the PTX loading per unit protein mass on HSA-PMEMA was approximately 1.43 times that of HSA. These collectively contributed to a 1.78-fold overall PTX intracellular delivery efficiency of HSA-PMEMA compared to that of HSA. Conclusions: In comparison with HSA, HSA-PMEMA nanomicelles exhibit improved cellular uptake and higher loading efficiency for PTX, effectively promoting the intracellular delivery of PTX. Tremendous potential lies in these micelles for developing safer and more efficient next-generation PTX formulations for tumor treatment.
Collapse
Affiliation(s)
- Yang Chen
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China;
| | - Shuang Liang
- Central Laboratory, NMPA Key Laboratory for Dental Materials, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Peking University School and Hospital of Stomatology, Beijing 100081, China; (S.L.); (B.C.); (F.J.)
| | - Binglin Chen
- Central Laboratory, NMPA Key Laboratory for Dental Materials, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Peking University School and Hospital of Stomatology, Beijing 100081, China; (S.L.); (B.C.); (F.J.)
| | - Fei Jiao
- Central Laboratory, NMPA Key Laboratory for Dental Materials, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Peking University School and Hospital of Stomatology, Beijing 100081, China; (S.L.); (B.C.); (F.J.)
| | - Xuliang Deng
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China;
- Central Laboratory, NMPA Key Laboratory for Dental Materials, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Peking University School and Hospital of Stomatology, Beijing 100081, China; (S.L.); (B.C.); (F.J.)
| | - Xinyu Liu
- Central Laboratory, NMPA Key Laboratory for Dental Materials, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Peking University School and Hospital of Stomatology, Beijing 100081, China; (S.L.); (B.C.); (F.J.)
| |
Collapse
|
3
|
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
|
4
|
Marschner N, Haug N, Hegewisch-Becker S, Reiser M, Dörfel S, Lerchenmüller C, Linde H, Wolf T, Hof A, Kaiser-Osterhues A, Potthoff K, Jänicke M. Head-to-head comparison of treatment sequences in advanced pancreatic cancer-Real-world data from the prospective German TPK clinical cohort study. Int J Cancer 2024; 155:1629-1640. [PMID: 38956837 DOI: 10.1002/ijc.35071] [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: 12/20/2023] [Revised: 04/25/2024] [Accepted: 05/15/2024] [Indexed: 07/04/2024]
Abstract
There are no clear guidelines regarding the optimal treatment sequence for advanced pancreatic cancer, as head-to-head phase III randomised trials are missing. We assess real-world effectiveness of three common sequential treatment strategies by emulating a hypothetical randomised trial. This analysis included 1551 patients with advanced pancreatic cancer from the prospective, clinical cohort study Tumour Registry Pancreatic Cancer receiving FOLFIRINOX (n = 613) or gemcitabine/nab-paclitaxel (GEMNAB; n = 938) as palliative first-line treatment. We used marginal structural modelling to compare overall survival (OS) and time to deterioration (TTD) of health-related quality of life (HRQoL) between three common first- to second-line treatment sequences, adjusting for time-varying potential confounding. The sequences were: FOLFIRINOX→GEMNAB, GEMNAB→FOLFOX/OFF and GEMNAB→nanoliposomal irinotecan (NALIRI) + 5-fluorouracil. Outcome was also calculated stratified by patients' prognostic risk according to the Pancreatic Cancer Score. Median OS and TTD of HRQoL independent of risk were 10.7 [8.9, 11.9] and 6.4 [4.8, 7.7] months for FOLFIRINOX→GEMNAB, 8.4 [7.4, 9.7] and 5.8 [4.6, 7.1] months for GEMNAB→FOLFOX/OFF and 8.9 [7.8, 10.4] and 4.6 [4.1, 6.1] months for GEMNAB→NALIRI+5-fluorouracil. Compared to FOLFIRINOX→GEMNAB, OS and TTD were worse for poor-risk patients with GEMNAB→FOLFOX/OFF (OS: HR 2.09 [1.47, 2.98]; TTD: HR 1.97 [1.19, 3.27]) and those with GEMNAB→NALIRI+5-fluorouracil (OS: HR 1.35, [0.76, 2.39]; TTD: HR 2.62 [1.56, 4.42]). Brackets denote 95%-confidence intervals. The estimated real-world effectiveness of the three treatment sequences evaluated were largely comparable. Poor-risk patients might benefit from intensified treatment with FOLFIRINOX→GEMNAB in terms of clinical and patient-reported outcomes. Future randomised trials on sequential treatments in advanced pancreatic cancer are warranted.
Collapse
Affiliation(s)
- Norbert Marschner
- Med. Klinik 1, Universitätsklinik Freiburg, Freiburg, Germany
- iOMEDICO, Freiburg, Germany
| | - Nina Haug
- Biostatistics, iOMEDICO, Freiburg, Germany
| | | | - Marcel Reiser
- PIOH-Praxis Internistische Onkologie und Hämatologie, Köln, Germany
| | | | | | - Hartmut Linde
- MVZ für Blut- und Krebserkrankungen, Potsdam, Germany
| | - Thomas Wolf
- BAG, Gemeinschaftspraxis Hämatologie-Onkologie, Dresden, Germany
| | - Anna Hof
- Clinical Epidemiology and Health Economics, iOMEDICO, Freiburg, Germany
| | | | | | - Martina Jänicke
- Clinical Epidemiology and Health Economics, iOMEDICO, Freiburg, Germany
| |
Collapse
|
5
|
del Campo-Pedrosa R, Martín-Carnicero A, González-Marcos A, Martínez A. New model to predict survival in advanced pancreatic ductal adenocarcinoma patients by measuring GGT and LDH levels and monocyte count. Front Oncol 2024; 14:1411096. [PMID: 39435278 PMCID: PMC11491290 DOI: 10.3389/fonc.2024.1411096] [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: 04/02/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer with a poor survival outcome. Predicting patient survival allows physicians to tailor treatments to specific individuals. Thus, a simple and cost-effective prognosis model is sorely needed. Methods This retrospective study assesses the prognostic value of blood biomarkers in advanced and metastatic PDAC patients (n=96) from Spain. Cut-off points for hematological parameters were calculated and correlated with overall survival (OS) using Kaplan-Meier, log-rank test, robust Cox proportional hazards and logistic regressions. Results In univariate analysis, individuals with low levels of GGT, LDH, ALP, leukocyte-, neutrophil- and monocyte counts showed significantly longer survival than patients with higher levels. In multivariate analysis, lower levels of GGT (HR (95%CI), 2.734 (1.223-6.111); p=0.014), LDH (HR (95%CI), 1.876 (1.035-3.400); p=0.038) and monocyte count (HR (95%CI), 1.657 (1.095-2.506); p = 0.017) remained significantly beneficial. In consequence, we propose a prognostic model based on logistic regression (AUC=0.741) of these three biomarkers as a pioneer tool to estimate OS in PDAC. Conclusion This study has demonstrated that the joint use of GGT (<92.00), LDH (<220.00) and monocyte count (<800) are independent positive prognostic factors in PDAC that can predict one-year survival in a novel prognostic logistic model.
Collapse
Affiliation(s)
- Rocío del Campo-Pedrosa
- Data Department, Encore Lab S.L., Logroño, Spain
- Department of Mechanical Engineering, Universidad de La Rioja, Logroño, Spain
| | | | - Ana González-Marcos
- Department of Mechanical Engineering, Universidad de La Rioja, Logroño, Spain
| | - Alfredo Martínez
- Angiogenesis Group, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| |
Collapse
|
6
|
Boyd LN, Ali M, Puik JR, Comandatore A, Ginocchi L, Meijer LL, Swijnenburg RJ, Tartarini R, Le Large TY, Morelli L, Garajova I, Besselink MG, Mambrini A, Wilmink JW, Frampton AE, van Laarhoven HW, Giovannetti E, Kazemier G. Plasma miR-379 can predict treatment response to FOLFIRINOX and gemcitabine- nab-paclitaxel in advanced pancreatic cancer. THE JOURNAL OF LIQUID BIOPSY 2024; 5:100152. [PMID: 40027944 PMCID: PMC11863948 DOI: 10.1016/j.jlb.2024.100152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 03/05/2025]
Abstract
Background Novel biomarkers, such as plasma microRNAs (miRs), are needed to help guide clinical decision-making for the type of chemotherapy to use in patients with advanced pancreatic ductal adenocarcinoma (PDAC). This study assessed the ability of plasma miRs to predict optimal treatment response from FOLFIRINOX or gemcitabine-nab-paclitaxel in these patients. Methods Next-generation sequencing (NGS) was performed for biomarker discovery in pre-treatment plasma samples from advanced PDAC patients subsequently treated with FOLFIRINOX (n = 12) or gemcitabine-nab-paclitaxel (n = 12). Selected candidate biomarkers were validated in 40 patients with advanced PDAC using RT-qPCR. Cox regression was then used to assess the predictive value of plasma miRs for either FOLFIRINOX or gemcitabine-nab-paclitaxel. Results In the validation cohort, high plasma miR-379 expression was strongly predictive of treatment response (Pinteraction = 0.0004). Overall survival (OS) was significantly better with FOLFIRINOX vs. gemcitabine-nab-paclitaxel in those patients with lower plasma miR-379 expression (hazard ratio, 0.32 [95% confidence interval, 0.08 to 0.98]; P = 0.046). However, gemcitabine-nab-paclitaxel was associated with superior OS in patients with higher plasma miR-379 (hazard ratio, 0.28 [0.10 to 0.86]; P = 0.027). In contrast, miR-127, miR-155, and miR-200 showed no predictive value for treatment response for either chemotherapy regimen (P interaction = 0.12, P interaction = 0.83 and P interaction = 0.12, respectively). Conclusions Plasma miR-379 appears clinically useful as a predictive biomarker to identify which patients with advanced PDAC benefit most from treatment with FOLFIRINOX or gemcitabine-nab-paclitaxel. Further validation in larger studies and clinical trials is now warranted.
Collapse
Affiliation(s)
- Lenka N.C. Boyd
- Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, the Netherlands
- Amsterdam UMC, Location Vrije Universiteit, Department of Medical Oncology, Lab of Medical Oncology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Mahsoem Ali
- Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, the Netherlands
- Amsterdam UMC, Location Vrije Universiteit, Department of Medical Oncology, Lab of Medical Oncology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Jisce R. Puik
- Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, the Netherlands
- Amsterdam UMC, Location Vrije Universiteit, Department of Medical Oncology, Lab of Medical Oncology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Annalisa Comandatore
- Amsterdam UMC, Location Vrije Universiteit, Department of Medical Oncology, Lab of Medical Oncology, Amsterdam, the Netherlands
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Laura Ginocchi
- Department of Oncology, Azienda Sanitaria Locale Toscana Nord Ovest, Massa Carrara Hospital, Massa, Italy
| | - Laura L. Meijer
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Rutger-Jan Swijnenburg
- Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, the Netherlands
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands
| | - Roberta Tartarini
- Department of Oncology, Azienda Sanitaria Locale Toscana Nord Ovest, Massa Carrara Hospital, Massa, Italy
| | - Tessa Y.S. Le Large
- Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Ingrid Garajova
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Marc G. Besselink
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands
| | - Andrea Mambrini
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Johanna W. Wilmink
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands
| | - Adam E. Frampton
- HPB Surgical Unit, Royal Surrey County Hospital, Guildford, Surrey, UK
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, W120HS, London, UK
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, The Leggett Building, University of Surrey, Guildford, Surrey, GU2 7WG, UK
| | - Hanneke W.M. van Laarhoven
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands
| | - Elisa Giovannetti
- Amsterdam UMC, Location Vrije Universiteit, Department of Medical Oncology, Lab of Medical Oncology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisa per la Scienza, Pisa, Italy
| | - Geert Kazemier
- Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
7
|
Heervä E, Väliaho V, Nurmi H, Lietzen E, Ålgars A, Kauhanen S. Outcomes After Multimodality Treatment of Pancreatic Cancer in an Unselected Single-Center Cohort. Cancer Manag Res 2024; 16:1065-1076. [PMID: 39220815 PMCID: PMC11363961 DOI: 10.2147/cmar.s465512] [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: 04/08/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) remains a lethal and rarely resectable malignancy. Here we explore the outcomes of surgery, as compared to definitive radiotherapy (dRT) or systemic therapy only in PDAC. Methods Pancreatic surgery and radiotherapy in Southwest Finland have been centralized to Turku University Hospital. Previously validated population-based electronic health records database was searched for all unselected PDAC patients from the years 2009-2019. Main outcome was median overall survival (mOS). Demographics, pathology, surgery, and oncological treatment data were collected. Results We identified 1006 patients with PDAC, 49% male, median age 71 years and 77% presenting with metastatic disease. In total, 405 patients were treated; 92 resected, 26 dRT without resection and 287 systemic therapy only. mOS was 34.6 months for resected, 26.7 months for dRT, and 7.5 months for systemic therapy patients. Among the 88 patients with locally advanced inoperable PDAC, dRT was independently associated with longer mOS (26.7 months) as compared to systemic therapy only (mOS 10.6 months). Among the 287 patients treated with systemic therapy only, combination chemotherapy was independently associated with longer mOS (11.6 months) as compared to gemcitabine-monotherapy (6.8 months). In patients progressing to second-line systemic treatment after gemcitabine failure, mOS was the same (5.0 months) with single or combination regimens. Conclusion Surgery remains the only curative approach for PDAC. In locally advanced PDAC, dRT was associated with longer survival as compared to systemic therapy only. Concerning first-line systemic therapy, our results support the use of combination chemotherapy over single-agent therapy.
Collapse
Affiliation(s)
- Eetu Heervä
- Department of Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Vesa Väliaho
- Department of Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Heidi Nurmi
- Department of Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Elina Lietzen
- Department of Digestive Surgery, Turku University Hospital and University of Turku, Turku, Finland
| | - Annika Ålgars
- Department of Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Saila Kauhanen
- Department of Digestive Surgery, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
8
|
Ferdous KU, Tesfay MZ, Cios A, Shelton RS, Hartupee C, Urbaniak A, Chamcheu JC, Mavros MN, Giorgakis E, Mustafa B, Simoes CC, Miousse IR, Basnakian AG, Moaven O, Post SR, Cannon MJ, Kelly T, Nagalo BM. Enhancing Neoadjuvant Virotherapy's Effectiveness by Targeting Stroma to Improve Resectability in Pancreatic Cancer. Biomedicines 2024; 12:1596. [PMID: 39062169 PMCID: PMC11275208 DOI: 10.3390/biomedicines12071596] [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: 06/24/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
About one-fourth of patients with pancreatic ductal adenocarcinoma (PDAC) are categorized as borderline resectable (BR) or locally advanced (LA). Chemotherapy and radiation therapy have not yielded the anticipated outcomes in curing patients with BR/LA PDAC. The surgical resection of these tumors presents challenges owing to the unpredictability of the resection margin, involvement of vasculature with the tumor, the likelihood of occult metastasis, a higher ratio of positive lymph nodes, and the relatively larger size of tumor nodules. Oncolytic virotherapy has shown promising activity in preclinical PDAC models. Unfortunately, the desmoplastic stroma within the PDAC tumor microenvironment establishes a barrier, hindering the infiltration of oncolytic viruses and various therapeutic drugs-such as antibodies, adoptive cell therapy agents, and chemotherapeutic agents-in reaching the tumor site. Recently, a growing emphasis has been placed on targeting major acellular components of tumor stroma, such as hyaluronic acid and collagen, to enhance drug penetration. Oncolytic viruses can be engineered to express proteolytic enzymes that cleave hyaluronic acid and collagen into smaller polypeptides, thereby softening the desmoplastic stroma, ultimately leading to increased viral distribution along with increased oncolysis and subsequent tumor size regression. This approach may offer new possibilities to improve the resectability of patients diagnosed with BR and LA PDAC.
Collapse
Affiliation(s)
- Khandoker Usran Ferdous
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Mulu Z. Tesfay
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Aleksandra Cios
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
| | - Randal S. Shelton
- College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Conner Hartupee
- Division of Surgical Oncology, Department of Surgery, Louisiana State University (LSU) Health, New Orleans, LA 70112, USA; (C.H.); (O.M.)
| | - Alicja Urbaniak
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.U.); (I.R.M.)
| | - Jean Christopher Chamcheu
- Department of Biological Sciences and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA;
- Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Michail N. Mavros
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
- College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Emmanouil Giorgakis
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Bahaa Mustafa
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Camila C. Simoes
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Isabelle R. Miousse
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.U.); (I.R.M.)
| | - Alexei G. Basnakian
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Central Arkansas Veterans Healthcare System, John L. McClellan Memorial VA Hospital, Little Rock, AR 72205, USA
| | - Omeed Moaven
- Division of Surgical Oncology, Department of Surgery, Louisiana State University (LSU) Health, New Orleans, LA 70112, USA; (C.H.); (O.M.)
- Department of Interdisciplinary Oncology, Louisiana Cancer Research Center, Louisiana State University (LSU) Health, New Orleans, LA 70112, USA
| | - Steven R. Post
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Martin J. Cannon
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Thomas Kelly
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Bolni Marius Nagalo
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| |
Collapse
|
9
|
Tang CY, Yang SH, Li CP, Su YY, Chiu SC, Bai LY, Shan YS, Chen LT, Chuang SC, Chan DC, Yen CJ, Peng CM, Chiu TJ, Chen YY, Chen JS, Chiang NJ, Chou WC. Impact of previous S-1 treatment on efficacy of liposomal irinotecan plus 5-fluorouracil and leucovorin in patients with metastatic pancreatic cancer. Pancreatology 2024; 24:600-607. [PMID: 38565467 DOI: 10.1016/j.pan.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 03/22/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND/OBJECTIVES Liposomal irinotecan plus 5-fluorouracil and leucovorin (nal-IRI + 5-FU/LV) provides survival benefits for metastatic pancreatic adenocarcinoma (mPDAC) refractory to gemcitabine-based treatment, mainly gemcitabine plus nab-paclitaxel (GA), in current practice. Gemcitabine plus S-1 (GS) is another commonly administered first-line regimen before nab-paclitaxel reimbursement; however, the efficacy and safety of nal-IRI + 5-FU/LV for mPDAC after failed GS treatment has not been reported and was therefore explored in this study. METHODS In total, 177 patients with mPDAC received first-line GS or GA treatment, followed by second-line nal-IRI + 5-FU/LV treatment (identified from a multicenter retrospective cohort in Taiwan from 2018 to 2020); 85 and 92 patients were allocated to the GS and GA groups, respectively. Overall survival (OS), time-to-treatment failure (TTF), and adverse events were compared between the two groups. RESULTS The baseline characteristics of the two groups were generally similar; however, a higher median age (67 versus 62 years, p < 0.001) and fewer liver metastases (52% versus 78%, p < 0.001) were observed in the GS versus GA group. The median OS was 15.0 and 15.9 months in the GS and GA groups, respectively (p = 0.58). The TTF (3.1 versus 2.8 months, p = 0.36) and OS (7.6 versus 6.7 months, p = 0.83) after nal-IRI treatment were similar between the two groups. More patients in the GS group developed mucositis during nal-IRI treatment (15% versus 4%, p = 0.02). CONCLUSIONS The efficacy of second-line nal-IRI +5-FU/LV treatment was unaffected by prior S-1 exposure. GS followed by nal-IRI treatment is an alternative treatment sequence for patients with mPDAC.
Collapse
Affiliation(s)
- Cheng-Yu Tang
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Hung Yang
- Department of Oncology, National Taiwan University Hospital and National Taiwan University, Taipei, Taiwan; Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chung-Pin Li
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Clinical Skills Training, Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yung-Yeh Su
- Department of Oncology, National Cheng Kung University Hospital, Taipei, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | | | - Li-Yuan Bai
- Division of Hematology-Oncology, Department of Internal Medicine, China Medical University Hospital and China Medical University, Taichung, Taiwan
| | - Yan-Shen Shan
- Division of General Surgery, Department of Surgery, National Cheng Kung University Hospital, Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan; Department of Internal Medicine, Kaohsiung Medical University Hospital and Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih-Chang Chuang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital and Kaohsiung Medical University, Kaohsiung, Taiwan
| | - De-Chuan Chan
- Division of General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Jui Yen
- Department of Oncology, National Cheng Kung University Hospital, Taipei, Taiwan
| | - Cheng-Ming Peng
- Department of Surgery, Chung Shan Medical University Hospital and Chung Shan Medical University, Taichung, Taiwan
| | - Tai-Jan Chiu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung, Taiwan
| | - Yen-Yang Chen
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung, Taiwan
| | - Jen-Shi Chen
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Nai-Jung Chiang
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Wen-Chi Chou
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
10
|
Smith S, Drummond K, Dowling A, Bennett I, Campbell D, Freilich R, Phillips C, Ahern E, Reeves S, Campbell R, Collins IM, Johns J, Dumas M, Hong W, Gibbs P, Gately L. Improving Clinical Registry Data Quality via Linkage With Survival Data From State-Based Population Registries. JCO Clin Cancer Inform 2024; 8:e2400025. [PMID: 38924710 DOI: 10.1200/cci.24.00025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/26/2024] [Accepted: 05/03/2024] [Indexed: 06/28/2024] Open
Abstract
PURPOSE Real-world data (RWD) collected on patients treated as part of routine clinical care form the basis of cancer clinical registries. Capturing accurate death data can be challenging, with inaccurate survival data potentially compromising the integrity of registry-based research. Here, we explore the utility of data linkage (DL) to state-based registries to enhance the capture of survival outcomes. METHODS We identified consecutive adult patients with brain tumors treated in the state of Victoria from the Brain Tumour Registry Australia: Innovation and Translation (BRAIN) database, who had no recorded date of death and no follow-up within the last 6 months. Full name and date of birth were used to match patients in the BRAIN registry with those in the Victorian Births, Deaths and Marriages (BDM) registry. Overall survival (OS) outcomes were compared pre- and post-DL. RESULTS Of the 7,346 clinical registry patients, 5,462 (74%) had no date of death and no follow-up recorded within the last 6 months. Of the 5,462 patients, 1,588 (29%) were matched with a date of death in BDM. Factors associated with an increased number of matches were poor prognosis tumors, older age, and social disadvantage. OS was significantly overestimated pre-DL compared with post-DL for the entire cohort (pre- v post-DL: hazard ratio, 1.43; P < .001; median, 29.9 months v 16.7 months) and for most individual tumor types. This finding was present independent of the tumor prognosis. CONCLUSION As revealed by linkage with BDM, a high proportion of patients in a brain cancer clinical registry had missing death data, contributed to by informative censoring, inflating OS calculations. DL to pertinent registries on an ongoing basis should be considered to ensure accurate reporting of survival data and interpretation of RWD outcomes.
Collapse
Affiliation(s)
- Samuel Smith
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Oncology, St Vincent's Hospital Melbourne, Fitzroy, VIC, Australia
| | - Kate Drummond
- University of Melbourne, Parkville, VIC, Australia
- Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Anthony Dowling
- Department of Medical Oncology, St Vincent's Hospital Melbourne, Fitzroy, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
| | - Iwan Bennett
- Department of Neurosurgery, Alfred Health, Prahran, VIC, Australia
| | - David Campbell
- Department of Medical Oncology, Barwon Health, Geelong, VIC, Australia
| | - Ronnie Freilich
- Department of Neurology, Cabrini Hospital, Malvern, VIC, Australia
| | - Claire Phillips
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Elizabeth Ahern
- Department of Medical Oncology, Monash Health, Clayton, VIC, Australia
| | - Simone Reeves
- Department of Radiation Oncology, Ballarat Austin Radiation Oncology Centre, Ballarat, VIC, Australia
| | - Robert Campbell
- Department of Medical Oncology, Bendigo Health, Bendigo, VIC, Australia
| | - Ian M Collins
- Department of Medical Oncology, South West Oncology, Warnambool, VIC, Australia
| | - Julie Johns
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Megan Dumas
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Wei Hong
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Peter Gibbs
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Lucy Gately
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Neurosurgery, Alfred Health, Prahran, VIC, Australia
| |
Collapse
|
11
|
Jang DK, Kim YA, Lee JW, Kim HJ, Lee YS, Chun JW, Lee JC, Woo SM, Hwang JH. The Trends and Outcomes of Initial Palliative Chemotherapy in Patients with Pancreatic Cancer in Korea Based on National Health Insurance Service Data. J Clin Med 2024; 13:3229. [PMID: 38892939 PMCID: PMC11172641 DOI: 10.3390/jcm13113229] [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: 04/22/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Background/Objectives: The survival rate of patients with pancreatic cancer (PC) has improved gradually since the introduction of FOLFIRINOX (FFX) and gemcitabine + albumin-bound paclitaxel (GnP) regimens. However, the trends and outcomes of initial palliative chemotherapy before and after the advent of these regimens and their contribution to survival rates are not well understood. This study aimed to investigate this in patients with PC in Korea using claims data from the National Health Insurance Service (NHIS). Methods: Patients diagnosed with PC who underwent initial palliative chemotherapy between 2007 and 2019 were identified from the NHIS database. Patient demographics, comorbidities, chemotherapy regimens, and survival rates were analyzed using follow-up data up to 2020. Results: In total, 14,760 patients (mean age, 63.78 ± 10.18 years; men, 59.19%) were enrolled. As initial palliative chemotherapy, 3823 patients (25.90%) received gemcitabine alone; 2779 (18.83%) received gemcitabine + erlotinib; 1948 (13.20%) received FFX; and 1767 (11.97%) received GnP. The median survival values were 15.00 months for FFX; 11.04 months for GnP; 8.40 months for gemcitabine alone; and 8.51 months for gemcitabine + erlotinib. The adjusted hazard ratio (aHR) for GnP vs. FFX was 1.291 (95% CI, 1.206-1.383) in the multivariate Cox regression analysis of mortality. Radiation therapy (aHR, 0.667; 95% CI, 0.612-0.728) and second-line chemotherapy (aHR, 0.639; 95% CI, 0.597-0.684) were significantly associated with improved survival. Conclusions: Our study found that first-line chemotherapy with FFX was associated with significantly longer survival than the other regimens, although caution is needed in interpreting the results.
Collapse
Affiliation(s)
- Dong Kee Jang
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul 07061, Republic of Korea;
| | - Young Ae Kim
- Division of Cancer Control & Policy, National Cancer Control Institute, National Cancer Center, Goyang 10408, Republic of Korea; (Y.A.K.); (J.W.L.); (H.-J.K.)
| | - Jang Won Lee
- Division of Cancer Control & Policy, National Cancer Control Institute, National Cancer Center, Goyang 10408, Republic of Korea; (Y.A.K.); (J.W.L.); (H.-J.K.)
| | - Hak-June Kim
- Division of Cancer Control & Policy, National Cancer Control Institute, National Cancer Center, Goyang 10408, Republic of Korea; (Y.A.K.); (J.W.L.); (H.-J.K.)
| | - Yoon Suk Lee
- Department of Internal Medicine, Inje University Ilsan Paik Hospital, Goyang 10380, Republic of Korea;
| | - Jung Won Chun
- Research Institute, Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic of Korea;
| | - Jong-Chan Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| | - Sang Myung Woo
- Research Institute, Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic of Korea;
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| |
Collapse
|
12
|
Mendis S, Lipton L, To YH, Ananda S, Michael M, McLachlan SA, Thomson B, Loveday B, Knowles B, Fox A, Nikfarjam M, Usatoff V, Shapiro J, Clarke K, Pattison S, Chee CE, Zielinski R, Wong R, Gibbs P, Lee B. Early onset pancreatic cancer-exploring contemporary treatment and outcomes using real-world data. Br J Cancer 2024; 130:1477-1484. [PMID: 38448752 PMCID: PMC11058801 DOI: 10.1038/s41416-024-02619-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Pancreatic cancer incidence is increasing in younger populations. Differences between early onset pancreatic cancer (EOPC) and later onset pancreatic cancer (LOPC), and how these should inform management warrant exploration in the contemporary setting. METHODS A prospectively collected multi-site dataset on consecutive pancreatic adenocarcinoma patients was interrogated. Patient, tumour, treatment, and outcome data were extracted for EOPC (≤50 years old) vs LOPC (>50 years old). RESULTS Of 1683 patients diagnosed between 2016 and 2022, 112 (6.7%) were EOPC. EOPC more frequently had the tail of pancreas tumours, earlier stage disease, surgical resection, and trended towards increased receipt of chemotherapy in the curative setting compared to LOPC. EOPC more frequently received 1st line chemotherapy, 2nd line chemotherapy, and chemoradiotherapy than LOPC in the palliative setting. Recurrence-free survival was improved for the tail of pancreas EOPC vs LOPC in the resected setting; overall survival was superior for EOPC compared to LOPC across the resected, locally advanced unresectable and metastatic settings. CONCLUSIONS EOPC remains a small proportion of pancreatic cancer diagnoses. The more favourable outcomes in EOPC suggest these younger patients are overall deriving benefits from increased treatment in the curative setting and increased therapy in the palliative setting.
Collapse
Affiliation(s)
- Shehara Mendis
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
| | | | - Yat Hang To
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Sumitra Ananda
- University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Michael Michael
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Sue-Anne McLachlan
- University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, St Vincent's Hospital, Fitzroy, VIC, Australia
| | - Benjamin Thomson
- Department of Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Benjamin Loveday
- Department of Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Brett Knowles
- Department of Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- Epworth Healthcare, Melbourne, VIC, Australia
| | - Adrian Fox
- Department of Hepatobiliary Surgery, St Vincent's Hospital, Fitzroy, VIC, Australia
| | - Mehrdad Nikfarjam
- University of Melbourne, Parkville, VIC, Australia
- Department of Hepatobiliary Surgery, Austin Health, Heidelberg, VIC, Australia
| | | | - Julia Shapiro
- Department of Medicine, Alfred Hospital, Prahran, VIC, Australia
| | - Kate Clarke
- Department of Medical Oncology, Wellington Hospital, Wellington, New Zealand
| | - Sharon Pattison
- Department of Medicine, Dunedin School of Medicine, University of Otago, Otago, New Zealand
| | - Cheng Ean Chee
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Rob Zielinski
- Department of Medical Oncology, Orange Hospital, Orange, NSW, Australia
- Department of Medical Oncology, Dubbo Base Hospital, Dubbo, NSW, Australia
- Department of Medical Oncology, Bathurst Base Hospital, West Bathurst, NSW, Australia
| | - Rachel Wong
- Epworth Healthcare, Melbourne, VIC, Australia
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
- Department of Medical Oncology, Eastern Health, Box Hill, VIC, Australia
| | - Peter Gibbs
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
| | - Belinda Lee
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- Department of Medical Oncology, Northern Hospital, Epping, VIC, Australia
| |
Collapse
|
13
|
Yimamu A, Li J, Zhang H, Liang L, Feng L, Wang Y, Zhou C, Li S, Gao Y. Computed tomography and guidelines-based human-machine fusion model for predicting resectability of the pancreatic cancer. J Gastroenterol Hepatol 2024; 39:399-409. [PMID: 37957952 DOI: 10.1111/jgh.16401] [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: 09/07/2023] [Revised: 10/04/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND AND AIM The study aims to develop a hybrid machine learning model for predicting resectability of the pancreatic cancer, which is based on computed tomography (CT) and National Comprehensive Cancer Network (NCCN) guidelines. METHOD We retrospectively studied 349 patients. One hundred seventy-one cases from Center 1 and 92 cases from Center 2 were used as the primary training cohort, and 66 cases from Center 3 and 20 cases from Center 4 were used as the independent test dataset. Semi-automatic module of ITK-SNAP software was used to assist CT image segmentation to obtain three-dimensional (3D) imaging region of interest (ROI). There were 788 handcrafted features extracted for 3D ROI using PyRadiomics. The optimal feature subset consists of three features screened by three feature selection methods as the input of the SVM to construct the conventional radiomics-based predictive model (cRad). 3D ROI was used to unify the resolution by 3D spline interpolation method for constructing the 3D tumor imaging tensor. Using 3D tumor image tensor as input, 3D kernelled support tensor machine-based predictive model (KSTM), and 3D ResNet-based deep learning predictive model (ResNet) were constructed. Multi-classifier fusion ML model is constructed by fusing cRad, KSTM, and ResNet using multi-classifier fusion strategy. Two experts with more than 10 years of clinical experience were invited to reevaluate each patient based on their CECT following the NCCN guidelines to obtain resectable, unresectable, and borderline resectable diagnoses. The three results were converted into probability values of 0.25, 0.75, and 0.50, respectively, according to the traditional empirical method. Then it is used as an independent classifier and integrated with multi-classifier fusion machine learning (ML) model to obtain the human-machine fusion ML model (HMfML). RESULTS Multi-classifier fusion ML model's area under receiver operating characteristic curve (AUC; 0.8610), predictive accuracy (ACC: 80.23%), sensitivity (SEN: 78.95%), and specificity (SPE: 80.60%) is better than cRad, KSTM, and ResNet-based single-classifier models and their two-classifier fusion models. This means that three different models have mined complementary CECT feature expression from different perspectives and can be integrated through CFS-ER, so that the fusion model has better performance. HMfML's AUC (0.8845), ACC (82.56%), SEN (84.21%), SPE (82.09%). This means that ML models might learn extra information from CECT that experts cannot distinguish, thus complementing expert experience and improving the performance of hybrid ML models. CONCLUSION HMfML can predict PC resectability with high accuracy.
Collapse
Affiliation(s)
- Adilijiang Yimamu
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Li
- School of Biomedical Engineering, Guangdong Provincial Key Laboratory of Medical Image Processing, Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
| | - Haojie Zhang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lidu Liang
- School of Biomedical Engineering, Guangdong Provincial Key Laboratory of Medical Image Processing, Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
| | - Lei Feng
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Wang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chenjie Zhou
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shulong Li
- School of Biomedical Engineering, Guangdong Provincial Key Laboratory of Medical Image Processing, Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
| | - Yi Gao
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Orlandi E, Citterio C, Chinelli R, Dotti I, Zaffignani E, Biasini C, Anselmi E, Cremona G, Vecchia S. Comparing Combination vs Monochemotherapy in Late-Elderly Patients With Advanced Pancreatic Cancer: Insights From a Single-Center Study. Cancer Control 2024; 31:10732748241304968. [PMID: 39620663 PMCID: PMC11613287 DOI: 10.1177/10732748241304968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/01/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
OBJECTIVES The incidence of pancreatic ductal adenocarcinoma (PDAC) increases with age and is frequently diagnosed at an inoperable stage, which limits treatment options. There is limited evidence concerning patients over 75 years old, and clinical practice often lacks clear guidance regarding the choice of first-line therapy. The primary objective of this retrospective study was to assess overall survival (OS) in elderly patients receiving first-line monochemotherapy vs combination therapy. Secondary objectives included evaluating progression-free survival (PFS) and safety. METHODS This retrospective study analyzed the records of 150 patients aged 75 or older with confirmed PDAC treated with first-line chemotherapy at Piacenza General Hospital, Italy. Of these, 72 patients received monotherapy, while 78 underwent polychemotherapy. The majority of patients (93.3%) were administered reduced doses, and within this group, 67.9% had their doses reduced by more than 20%. Most patients (80%) presented with comorbidities, predominantly hypertension and diabetes. RESULTS The median overall survival was significantly higher in the polychemotherapy group (8.2 months) compared to the monotherapy group (4.7 months), with a P-value of 0.0022. The median PFS was 5.7 months for polychemotherapy and 2.8 months for monotherapy, showing a statistically significant difference (P = 0.004). In the multivariate analysis, poor performance status, high CA19.9 levels, and monotherapy were significantly associated with worse OS. Patients treated with polychemotherapy had a 37% lower likelihood of death within the year compared to those treated with monotherapy (HR 0.58, P = 0.009). CONCLUSION Polychemotherapy may provide a survival advantage over monotherapy in the late-elderly population, although considerations for dose adjustments due to comorbidities and polypharmacy are necessary. These findings suggest that, when feasible, polychemotherapy could offer a balance between effectiveness and tolerability, potentially improving outcomes in this age group.
Collapse
Affiliation(s)
- Elena Orlandi
- Department of Oncology and Hematology, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Chiara Citterio
- Department of Oncology and Hematology, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Rebecca Chinelli
- Department of Pharmacy, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Ilaria Dotti
- Department of Pharmacy, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Elena Zaffignani
- Department of Oncology and Hematology, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Claudia Biasini
- Department of Oncology and Hematology, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Elisa Anselmi
- Department of Oncology and Hematology, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Gabriele Cremona
- Department of Oncology and Hematology, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Stefano Vecchia
- Department of Pharmacy, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| |
Collapse
|
15
|
Pedrazzoli S. Currently Debated Topics on Surgical Treatment of Pancreatic Ductal Adenocarcinoma: A Narrative Review on Surgical Treatment of Borderline Resectable, Locally Advanced, and Synchronous or Metachronous Oligometastatic Tumor. J Clin Med 2023; 12:6461. [PMID: 37892599 PMCID: PMC10607532 DOI: 10.3390/jcm12206461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/29/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Previously considered inoperable patients (borderline resectable, locally advanced, synchronous oligometastatic or metachronous pancreatic adenocarcinoma (PDAC)) are starting to become resectable thanks to advances in chemo/radiotherapy and the reduction in operative mortality. METHODS This narrative review presents a chosen literature selection, giving a picture of the current state of treatment of these patients. RESULTS Neoadjuvant therapy (NAT) is generally recognized as the treatment of choice before surgery. However, despite the increased efficacy, the best pathological response is still limited to 10.9-27.9% of patients. There are still limited data on the selection of possible NAT responders and how to diagnose non-responders early. Multidetector computed tomography has high sensitivity and low specificity in evaluating resectability after NAT, limiting the resection rate of resectable patients. Ca 19-9 and Positron emission tomography are giving promising results. The prediction of early recurrence after a radical resection of synchronous or metachronous metastatic PDAC, thus identifying patients with poor prognosis and saving them from a resection of little benefit, is still ongoing, although some promising data are available. CONCLUSION In conclusion, high-level evidence demonstrating the benefit of the surgical treatment of such patients is still lacking and should not be performed outside of high-volume centers with interdisciplinary teams of surgeons and oncologists.
Collapse
|
16
|
Merza N, Farooqui SK, Dar SH, Varughese T, Awan RU, Qureshi L, Ansari SA, Qureshi H, Mcilvaine J, Vohra I, Nawras Y, Kobeissy A, Hassan M. Folfirinox vs. Gemcitabine + Nab-Paclitaxel as the First-Line Treatment for Pancreatic Cancer: A Systematic Review and Meta-Analysis. World J Oncol 2023; 14:325-339. [PMID: 37869244 PMCID: PMC10588495 DOI: 10.14740/wjon1604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 07/07/2023] [Indexed: 10/24/2023] Open
Abstract
Background The efficacy and safety of Folfirinox (FFX) or gemcitabine + nab-paclitaxel (GnP) to be used as the first-line drugs for pancreatic cancer (PC) is yet to be established. We conducted an analysis of retrospective studies to assess the efficacy and safety of these two regimens by comparing their survival and safety outcomes in patients with PC. Methods We conducted an extensive review of two electronic databases from inception till February 2023 to include all the relevant studies that compared FFX with GnP published and unpublished work. Retrospective studies were only included. Overall survival (OS) and progression-free survival (PFS) were pooled using hazard ratios (HRs), while objective response rate (ORR) and safety outcomes were pooled using odds ratios (ORs) with 95% confidence interval (CI) using the random effects model. Results A total of 7,030 patients were identified in a total of 21 articles that were shortlisted. Pooled results concluded that neither FFX nor GnP was associated to increase the OS time (HR: 0.93, 95% CI: 0.83 - 1.04; P = 0.0001); however, FFX was more likely associated with increased PFS when compared to GnP (HR: 0.88, 95% CI: 0.81 - 0.97; P < 0.0001). ORR proved to be non-significant between the two regimens (OR: 0.90, 95% CI: 0.64 - 1.27; P = 0.15). Safety outcomes included neutropenia, anemia, thrombocytopenia and diarrhea. GnP was more associated with diarrhea (OR: 1.96, 95% CI: 1.22 - 3.15; P = 0.001), while FFX was seen to cause anemia (OR: 0.70, 95% CI: 0.51 - 0.98; P = 0.10) in PC patients. Neutropenia and thrombocytopenia were in-significant in the two drug regimens (OR: 1.10, 95% CI: 0.92 - 1.31; P = 0.33 and OR: 0.83, 95% CI: 0.60 - 1.13; P = 0.23, respectively). Conclusion FFX and GnP showed a significant difference in increasing the PFS, while no difference was observed while measuring OS. Safety outcomes showed that FFX and GnP shared similar safety profiles as FFX was associated with hematological outcomes, while GnP was more associated with non-hematological outcomes.
Collapse
Affiliation(s)
- Nooraldin Merza
- Department of Internal Medicine, University of Toledo, Toledo, OH, USA
| | | | - Sophia Haroon Dar
- Department of Internal Medicine, Long Island Jewish Medical Center-Northshore University Hospital, Manhasset, NY, USA
| | - Tony Varughese
- Department of Internal Medicine, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Rehmat Ullah Awan
- Department of Internal Medicine, Ochsner Rush Hospital, Meridian, MS, USA
| | - Lamaan Qureshi
- Edson College of Nursing and Health Innovations, Tempe, AZ, USA
| | - Saad Ali Ansari
- Department of Internal Medicine, University of California, Riverside School of Medicine, Riverside, CA, USA
| | - Hadi Qureshi
- School of Liberal Arts, Arizona State University, Maricopa, AZ, USA
| | - Jamie Mcilvaine
- Department of OBGYN-Rutgers Jersey City, Jersey City, NJ, USA
| | - Ishaan Vohra
- Gastroenterology Department, University of Illinois, Peoria, IL, USA
| | - Yusuf Nawras
- University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Abdallah Kobeissy
- Department of Gastroenterology, University of Toledo, Toledo, OH, USA
| | - Mona Hassan
- Department of Gastroenterology, University of Toledo, Toledo, OH, USA
| |
Collapse
|
17
|
Urbanova M, Cihova M, Buocikova V, Slopovsky J, Dubovan P, Pindak D, Tomas M, García-Bermejo L, Rodríguez-Garrote M, Earl J, Kohl Y, Kataki A, Dusinska M, Sainz B, Smolkova B, Gabelova A. Nanomedicine and epigenetics: New alliances to increase the odds in pancreatic cancer survival. Biomed Pharmacother 2023; 165:115179. [PMID: 37481927 DOI: 10.1016/j.biopha.2023.115179] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest cancers worldwide, primarily due to its robust desmoplastic stroma and immunosuppressive tumor microenvironment (TME), which facilitate tumor progression and metastasis. In addition, fibrous tissue leads to sparse vasculature, high interstitial fluid pressure, and hypoxia, thereby hindering effective systemic drug delivery and immune cell infiltration. Thus, remodeling the TME to enhance tumor perfusion, increase drug retention, and reverse immunosuppression has become a key therapeutic strategy. In recent years, targeting epigenetic pathways has emerged as a promising approach to overcome tumor immunosuppression and cancer progression. Moreover, the progress in nanotechnology has provided new opportunities for enhancing the efficacy of conventional and epigenetic drugs. Nano-based drug delivery systems (NDDSs) offer several advantages, including improved drug pharmacokinetics, enhanced tumor penetration, and reduced systemic toxicity. Smart NDDSs enable precise targeting of stromal components and augment the effectiveness of immunotherapy through multiple drug delivery options. This review offers an overview of the latest nano-based approaches developed to achieve superior therapeutic efficacy and overcome drug resistance. We specifically focus on the TME and epigenetic-targeted therapies in the context of PDAC, discussing the advantages and limitations of current strategies while highlighting promising new developments. By emphasizing the immense potential of NDDSs in improving therapeutic outcomes in PDAC, our review paves the way for future research in this rapidly evolving field.
Collapse
Affiliation(s)
- Maria Urbanova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Marina Cihova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Verona Buocikova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Jan Slopovsky
- 2nd Department of Oncology, National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia; Faculty of Medicine, Comenius University, Spitalska 24, 813 72 Bratislava, Slovakia
| | - Peter Dubovan
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia; Department of Surgical Oncology, National CancerInstitute in Bratislava, Klenova 1, 833 10 Bratislava, Slovakia; Faculty of Medicine, Slovak Medical University in Bratislava, Limbová12, 833 03 Bratislava
| | - Daniel Pindak
- Department of Surgical Oncology, National CancerInstitute in Bratislava, Klenova 1, 833 10 Bratislava, Slovakia; Faculty of Medicine, Slovak Medical University in Bratislava, Limbová12, 833 03 Bratislava
| | - Miroslav Tomas
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia; Department of Surgical Oncology, National CancerInstitute in Bratislava, Klenova 1, 833 10 Bratislava, Slovakia; Faculty of Medicine, Slovak Medical University in Bratislava, Limbová12, 833 03 Bratislava
| | - Laura García-Bermejo
- Biomarkers and Therapeutic Targets Group, Area4, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain
| | - Mercedes Rodríguez-Garrote
- Molecular Epidemiology and Predictive Tumor Markers Group, Area 3, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain; CIBERONC, Madrid, Spain
| | - Julie Earl
- Molecular Epidemiology and Predictive Tumor Markers Group, Area 3, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain; CIBERONC, Madrid, Spain
| | - Yvonne Kohl
- Department Bioprocessing & Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, 66280 Sulzbach, Germany
| | - Agapi Kataki
- 1st Department of Propaedeutic Surgery, National and Kapodistrian University of Athens, Vasilissis Sofias 114, 11527 Athens, Greece
| | - Maria Dusinska
- Health Effects Laboratory, Department of Environmental Chemistry, NILU-Norwegian Institute for Air Research, Instituttveien 18, 2002 Kjeller, Norway
| | - Bruno Sainz
- CIBERONC, Madrid, Spain; Instituto de Investigaciones Biomédicas"Alberto Sols" (IIBM), CSIC-UAM, 28029 Madrid, Spain; Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Area 3, Ramón y Cajal Health Research Institute (IRYCIS), 28034 Madrid, Spain
| | - Bozena Smolkova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Alena Gabelova
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 84505 Bratislava, Slovakia..
| |
Collapse
|
18
|
Ying N, Liu S, Zhang M, Cheng J, Luo L, Jiang J, Shi G, Wu S, Ji J, Su H, Pan H, Zeng D. Nano delivery system for paclitaxel: Recent advances in cancer theranostics. Colloids Surf B Biointerfaces 2023; 228:113419. [PMID: 37393700 DOI: 10.1016/j.colsurfb.2023.113419] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/22/2023] [Accepted: 06/17/2023] [Indexed: 07/04/2023]
Abstract
Paclitaxel is one of the most effective chemotherapeutic drugs which processes the obvious curative effect for a broad range of cancers including breast, ovarian, lung, and head & neck cancers. Though some novel paclitaxel-loaded formulations have been developed, the clinical application of the paclitaxel is still limited due to its toxicity and solubility issues. Over the past decades, we have seen rapid advances in applying nanocarriers in paclitaxel delivery systems. The nano-drug delivery systems offer unique advantages in enhancing the aqueous solubility, reducing side effects, increasing permeability, prolonging circulation half-life of paclitaxel. In this review, we summarize recent advances in developing novel paclitaxel-loaded nano delivery systems based on nanocarriers. These nanocarriers show great potentials in overcoming the disadvantages of pure paclitaxel and as a result improving the efficacy.
Collapse
Affiliation(s)
- Na Ying
- Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Sisi Liu
- Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mengmeng Zhang
- Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jing Cheng
- Shanghai University of Medicine & Health Sciences, Shanghai 201318, China
| | - Linghuan Luo
- Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Jiayi Jiang
- Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Gaofan Shi
- Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Shu Wu
- Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Jun Ji
- Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Haoyuan Su
- Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Hongzhi Pan
- Shanghai University of Medicine & Health Sciences, Shanghai 201318, China.
| | - Dongdong Zeng
- Shanghai University of Medicine & Health Sciences, Shanghai 201318, China.
| |
Collapse
|
19
|
Lee JH, Lee SH, Lee SK, Choi JH, Lim S, Kim MS, Lee KM, Lee MW, Ku JL, Kim DH, Cho IR, Paik WH, Ryu JK, Kim YT. Antiproliferative Activity of Krukovine by Regulating Transmembrane Protein 139 (TMEM139) in Oxaliplatin-Resistant Pancreatic Cancer Cells. Cancers (Basel) 2023; 15:cancers15092642. [PMID: 37174108 PMCID: PMC10177337 DOI: 10.3390/cancers15092642] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/24/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Krukovine (KV) is an alkaloid isolated from the bark of Abuta grandifolia (Mart.) Sandw. (Menispermaceae) with anticancer potential in some cancers with KRAS mutations. In this study, we explored the anticancer efficacy and mechanism of KV in oxaliplatin-resistant pancreatic cancer cells and patient-derived pancreatic cancer organoids (PDPCOs) with KRAS mutation. After treatment with KV, mRNA and protein levels were determined by RNA-seq and Western blotting, respectively. Cell proliferation, migration, and invasion were measured by MTT, scratch wound healing assay, and transwell analysis, respectively. Patient-derived pancreatic cancer organoids (PDPCOs) with KRAS mutations were treated with KV, oxaliplatin (OXA), and a combination of KV and OXA. KV suppresses tumor progression via the downregulation of the Erk-RPS6K-TMEM139 and PI3K-Akt-mTOR pathways in oxaliplatin-resistant AsPC-1 cells. Furthermore, KV showed an antiproliferative effect in PDPCOs, and the combination of OXA and KV inhibited PDPCO growth more effectively than either drug alone.
Collapse
Affiliation(s)
- Jee-Hyung Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Natural Products Research Institute, Seoul National University College of Pharmacy, Seoul 08826, Republic of Korea
| | - Sang-Hyub Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sang-Kook Lee
- Natural Products Research Institute, Seoul National University College of Pharmacy, Seoul 08826, Republic of Korea
| | - Jin-Ho Choi
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea
| | - Seohyun Lim
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Min-Song Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Kyung-Min Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Min-Woo Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Ja-Lok Ku
- Department of Biomedical Sciences, Korean Cell Line Bank, Laboratory of Cell Biology and Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Dae-Hyun Kim
- Dxome Co., Ltd., Seongnam 331, Republic of Korea
| | - In-Rae Cho
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Woo-Hyun Paik
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Ji-Kon Ryu
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Yong-Tae Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|