1
|
Martin RC, Li Y, Shore EA, Malik DA, Li H, Hu X, Hayat T, Tan M, McMasters KM, Yan J. Irreversible Electroporation and Beta-Glucan-Induced Trained Innate Immunity for Treatment of Pancreatic Ductal Adenocarcinoma: A Phase II Study. J Am Coll Surg 2025; 240:351-361. [PMID: 39840846 PMCID: PMC11928255 DOI: 10.1097/xcs.0000000000001291] [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] [Indexed: 01/23/2025]
Abstract
BACKGROUND Irreversible electroporation (IRE) has augmented the effects of certain immunotherapies in pancreatic ductal adenocarcinoma (PDA). Yeast-derived particulate beta-glucan induces trained innate immunity and successfully reduced murine pancreatic cancer burden. This is a phase II study to test the hypothesis that IRE may augment beta-glucan-induced trained immunity in patients with PDA. STUDY DESIGN In this phase II clinical trial (NCT03080974), surgical ablative IRE was performed on clinical stage III PDA followed by oral beta-glucan administration for 12 months or until disease recurrence. Peripheral blood was taken preoperative, 14 days, and every 3 months and was evaluated by mass cytometry and compared with patients who received IRE alone. RESULTS Thirty consecutive patients with preoperative clinical stage III PDA were treated with IRE and then initiated on oral beta-glucan postoperatively were compared with 20 patients treated with IRE alone. There were no dose-limiting toxicities with oral beta-glucan, and compliance with therapy was 96% in all patients. Seven patients (23%) developed grade 3 or 4 treatment-related adverse events at 90 days; none required a dose modification of oral beta-glucan. A median disease-free interval (DFI) was 18 months (range 6 to 48 months), with a median overall survival (OS) of 32.5 months (range 4 to 53 months). At 12 months post-IRE, immunophenotyping was demonstrated a significant effect with improvement in the IRE-beta-glucan-treated group. This also resulted in a significant decrease on naive CD4 and CD8 T cells with increased CD4 and CD8 terminal effector cells in the IRE-beta-glucan-treated group, which correlated with a significant improvement in DFI and OS (p = 0.001). CONCLUSIONS Combined beta-glucan with IRE-ablated PDA tumor cells elicited a potent trained response and augmented antitumor functionality at 12 months post-IRE, which translated into an improved DFI and OS.
Collapse
Affiliation(s)
- Robert C.G. Martin
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | - Yan Li
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | - Emily A. Shore
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | - Danial A Malik
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | - Hong Li
- Functional Immunomics Core, Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Xiaoling Hu
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Traci Hayat
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | - Min Tan
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | - Kelly M. McMasters
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jun Yan
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
2
|
Kataki AD, Gupta PG, Cheema U, Nisbet A, Wang Y, Kocher HM, Pérez-Mancera PA, Velliou EG. Mapping Tumor-Stroma-ECM Interactions in Spatially Advanced 3D Models of Pancreatic Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:16708-16724. [PMID: 40052705 PMCID: PMC11931495 DOI: 10.1021/acsami.5c02296] [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] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/21/2025]
Abstract
Bioengineering-based in vitro tumor models are increasingly important as tools for studying disease progression and therapy response for many cancers, including the deadly pancreatic ductal adenocarcinoma (PDAC) that exhibits a tumor/tissue microenvironment of high cellular/biochemical complexity. Therefore, it is crucial for in vitro models to capture that complexity and to enable investigation of the interplay between cancer cells and factors such as extracellular matrix (ECM) proteins or stroma cells. Using polyurethane (PU) scaffolds, we performed a systematic study on how different ECM protein scaffold coatings impact the long-term cell evolution in scaffolds containing only cancer or only stroma cells (activated stellate and endothelial cells). To investigate potential further changes in those biomarkers due to cancer-stroma interactions, we mapped their expression in dual/zonal scaffolds consisting of a cancer core and a stroma periphery, spatially mimicking the fibrotic/desmoplastic reaction in PDAC. In our single scaffolds, we observed that the protein coating affected the cancer cell spatial aggregation, matrix deposition, and biomarker upregulation in a cell-line-dependent manner. In single stroma scaffolds, different levels of fibrosis/desmoplasia in terms of ECM composition/quantity were generated depending on the ECM coating. When studying the evolution of cancer and stroma cells in our dual/zonal model, biomarkers linked to cell aggressiveness/invasiveness were further upregulated by both cancer and stroma cells as compared to single scaffold models. Collectively, our study advances the understanding of how different ECM proteins impact the long-term cell evolution in PU scaffolds. Our findings show that within our bioengineered models, we can stimulate the cells of the PDAC microenvironment to develop different levels of aggressiveness/invasiveness, as well as different levels of fibrosis. Furthermore, we highlight the importance of considering spatial complexity to map cell invasion. Our work contributes to the design of in vitro models with variable, yet biomimetic, tissue-like properties for studying the tumor microenvironment's role in cancer progression.
Collapse
Affiliation(s)
- Anna-Dimitra Kataki
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
| | - Priyanka G. Gupta
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
- School
of Life and Health Sciences, Whitelands College, University of Roehampton, London SW15 4JD, U.K.
| | - Umber Cheema
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
| | - Andrew Nisbet
- Department
of Medical Physics and Biomedical Engineering, University College London, London WC1E 6BT, U.K.
| | - Yaohe Wang
- Centre
for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, U.K.
| | - Hemant M. Kocher
- Centre
for Tumour Biology and Experimental Cancer Medicine, Barts Cancer
Institute, Queen Mary University of London, London EC1M 6BQ, U.K.
| | - Pedro A. Pérez-Mancera
- Department
of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GE, U.K.
| | - Eirini G. Velliou
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
| |
Collapse
|
3
|
Chihanga T, Xu S, Fultz HN, Nicholson JD, Brombacher MD, Hawkins K, Fay DR, Steil MM, Ni S, Kennedy MA. How Early Can Pancreatic Tumors Be Detected Using NMR-Based Urine Metabolic Profiling? Identification of Early-Stage Biomarkers of Tumor Initiation and Progression in an Orthotopic Xenograft Mouse Model of Pancreatic Cancer. Metabolites 2025; 15:142. [PMID: 40137107 PMCID: PMC11943925 DOI: 10.3390/metabo15030142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 01/29/2025] [Accepted: 02/02/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Pancreatic cancer is the most lethal of all human cancers. The disease has no obvious symptoms in its early stages and in the majority of cases, the cancer goes undetected until it has advanced to the point that surgery is no longer a viable option or until it has metastasized to other organs. The absence of reliable and sensitive biomarkers for the early detection of pancreatic cancer contributes to the poor ability to detect the disease before it progresses to an untreatable stage. Objectives: Here, an orthotopic xenograft mouse model of pancreatic cancer was investigated to determine if urinary metabolic biomarkers could be identified and used to detect the early formation of pancreatic tumors. Methods: The orthotopic xenograft mouse model of pancreatic cancer was established by injecting human MiaPaCa-2 cells, derived from a male patient aged 65 years with pancreatic adenocarcinoma, into the pancreata of severe combined immunodeficient mice. Orthotopic pancreatic tumors, allowed to grow for eight weeks, were successfully established in the pancreata in 15 out of 20 mice. At the time of sacrifice, tumors were excised and histologically analyzed and the masses and volumes recorded. Urine samples were collected prior to injection, at one-week post injection, and every two weeks afterwards for eight weeks. Results: NMR-based metabolic profiling of the urine samples indicated that 31 metabolites changed significantly over the course of tumor initiation and growth. Longitudinal metabolic profiling analysis indicated an initial increase in activity of the metabolic pathways involved in energy production and/or cell synthesis by cancer cells as required to support tumor growth that was followed by a diminished difference between control and orthotopic mice associated with tumor senescence as the tumors reached 7-8 weeks post injection. Conclusions: The results indicate that NMR-based urinary metabolic profiling may be able to detect the earliest stages of pancreatic tumor initiation and growth, highlighting the potential for translation to human clinical studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Michael A. Kennedy
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH 45056, USA; (T.C.); (S.X.); (H.N.F.); (J.D.N.); (M.D.B.); (K.H.); (D.R.F.); (S.N.)
| |
Collapse
|
4
|
Alam MS, Gaida MM, Witzel HR, Otsuka S, Abbasi A, Guerin T, Abdelmaksoud A, Wong N, Cam MC, Kozlov S, Ashwell JD. TNFR1 signaling promotes pancreatic tumor growth by limiting dendritic cell number and function. Cell Rep Med 2024; 5:101696. [PMID: 39178856 PMCID: PMC11528236 DOI: 10.1016/j.xcrm.2024.101696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/28/2024] [Accepted: 07/30/2024] [Indexed: 08/26/2024]
Abstract
Pancreatic adenocarcinoma (PDAC) is one the most intractable cancers, in part due to its highly inflammatory microenvironment and paucity of infiltrating dendritic cells (DCs). Here, we find that genetic ablation or antibody blockade of tumor necrosis factor receptor 1 (TNFR1) enhanced intratumor T cell activation and slowed PDAC growth. While anti-PD-1 checkpoint inhibition alone had little effect, it further enhanced intratumor T cell activation in combination with anti-TNFR1. The major cellular alteration in the tumor microenvironment in the absence of TNFR1 signaling was a large increase in DC number and immunostimulatory phenotype. This may reflect a direct effect on DCs, because TNF induced TNFR1-dependent apoptosis of bone-marrow-derived DCs. The therapeutic response to anti-TNFR1 alone was superior to the combination of DC-activating agonistic anti-CD40 and Flt3 ligand (Flt3L). These observations suggest that targeting TNFR1, perhaps in concert with other strategies that promote DC generation and mobilization, may have therapeutic benefits.
Collapse
Affiliation(s)
- Muhammad S Alam
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Matthias M Gaida
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; TRON, Translational Oncology at the University Medical Center, JGU-Mainz, 55131 Mainz, Germany; Research Center for Immunotherapy, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany
| | - Hagen R Witzel
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany
| | - Shizuka Otsuka
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aamna Abbasi
- Department of Integrative Immunobiology, Duke University, Durham, NC 27708, USA
| | - Theresa Guerin
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21707, USA
| | - Abdalla Abdelmaksoud
- Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Nathan Wong
- Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Margaret C Cam
- Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Serguei Kozlov
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21707, USA
| | - Jonathan D Ashwell
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
5
|
Plaugher D, Murrugarra D. Cancer mutationscape: revealing the link between modular restructuring and intervention efficacy among mutations. NPJ Syst Biol Appl 2024; 10:74. [PMID: 39003264 PMCID: PMC11246485 DOI: 10.1038/s41540-024-00398-6] [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: 02/12/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024] Open
Abstract
There is increasing evidence that biological systems are modular in both structure and function. Complex biological signaling networks such as gene regulatory networks (GRNs) are proving to be composed of subcategories that are interconnected and hierarchically ranked. These networks contain highly dynamic processes that ultimately dictate cellular function over time, as well as influence phenotypic fate transitions. In this work, we use a stochastic multicellular signaling network of pancreatic cancer (PC) to show that the variance in topological rankings of the most phenotypically influential modules implies a strong relationship between structure and function. We further show that induction of mutations alters the modular structure, which analogously influences the aggression and controllability of the disease in silico. We finally present evidence that the impact and location of mutations with respect to PC modular structure directly corresponds to the efficacy of single agent treatments in silico, because topologically deep mutations require deep targets for control.
Collapse
Affiliation(s)
- Daniel Plaugher
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA.
| | - David Murrugarra
- Department of Mathematics, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
6
|
Gupta P, Bermejo-Rodriguez C, Kocher H, Pérez-Mancera PA, Velliou EG. Chemotherapy Assessment in Advanced Multicellular 3D Models of Pancreatic Cancer: Unravelling the Importance of Spatiotemporal Mimicry of the Tumor Microenvironment. Adv Biol (Weinh) 2024; 8:e2300580. [PMID: 38327154 DOI: 10.1002/adbi.202300580] [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: 11/03/2023] [Revised: 01/10/2024] [Indexed: 02/09/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a challenge for global health with very low survival rate and high therapeutic resistance. Hence, advanced preclinical models for treatment screening are of paramount importance. Herein, chemotherapeutic (gemcitabine) assessment on novel (polyurethane) scaffold-based spatially advanced 3D multicellular PDAC models is carried out. Through comprehensive image-based analysis at the protein level, and expression analysis at the mRNA level, the importance of stromal cells is confirmed, primarily activated stellate cells in the chemoresistance of PDAC cells within the models. Furthermore, it is demonstrated that, in addition to the presence of activated stellate cells, the spatial architecture of the scaffolds, i.e., segregation/compartmentalization of the cancer and stromal zones, affect the cellular evolution and is necessary for the development of chemoresistance. These results highlight that, further to multicellularity, mapping the tumor structure/architecture and zonal complexity in 3D cancer models is important for better mimicry of the in vivo therapeutic response.
Collapse
Affiliation(s)
- Priyanka Gupta
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London, London, W1W 7TY, UK
| | - Camino Bermejo-Rodriguez
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Hemant Kocher
- Centre for Tumour Biology and Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Pedro A Pérez-Mancera
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Eirini G Velliou
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London, London, W1W 7TY, UK
| |
Collapse
|
7
|
Farhangnia P, Khorramdelazad H, Nickho H, Delbandi AA. Current and future immunotherapeutic approaches in pancreatic cancer treatment. J Hematol Oncol 2024; 17:40. [PMID: 38835055 PMCID: PMC11151541 DOI: 10.1186/s13045-024-01561-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024] Open
Abstract
Pancreatic cancer is a major cause of cancer-related death, but despondently, the outlook and prognosis for this resistant type of tumor have remained grim for a long time. Currently, it is extremely challenging to prevent or detect it early enough for effective treatment because patients rarely exhibit symptoms and there are no reliable indicators for detection. Most patients have advanced or spreading cancer that is difficult to treat, and treatments like chemotherapy and radiotherapy can only slightly prolong their life by a few months. Immunotherapy has revolutionized the treatment of pancreatic cancer, yet its effectiveness is limited by the tumor's immunosuppressive and hard-to-reach microenvironment. First, this article explains the immunosuppressive microenvironment of pancreatic cancer and highlights a wide range of immunotherapy options, including therapies involving oncolytic viruses, modified T cells (T-cell receptor [TCR]-engineered and chimeric antigen receptor [CAR] T-cell therapy), CAR natural killer cell therapy, cytokine-induced killer cells, immune checkpoint inhibitors, immunomodulators, cancer vaccines, and strategies targeting myeloid cells in the context of contemporary knowledge and future trends. Lastly, it discusses the main challenges ahead of pancreatic cancer immunotherapy.
Collapse
Affiliation(s)
- Pooya Farhangnia
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hamid Nickho
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Delbandi
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Plaugher D, Murrugarra D. Pancreatic cancer mutationscape: revealing the link between modular restructuring and intervention efficacy amidst common mutations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.27.577546. [PMID: 38352601 PMCID: PMC10862704 DOI: 10.1101/2024.01.27.577546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/04/2024]
Abstract
There is increasing evidence that biological systems are modular in both structure and function. Complex biological signaling networks such as gene regulatory networks (GRNs) are proving to be composed of subcategories that are interconnected and hierarchically ranked. These networks contain highly dynamic processes that ultimately dictate cellular function over time, as well as influence phenotypic fate transitions. In this work, we use a stochastic multicellular signaling network of pancreatic cancer (PC) to show that the variance in topological rankings of the most phenotypically influential modules implies a strong relationship between structure and function. We further show that induction of mutations alters the modular structure, which analogously influences the aggression and controllability of the disease in silico. We finally present evidence that the impact and location of mutations with respect to PC modular structure directly corresponds to the efficacy of single agent treatments in silico, because topologically deep mutations require deep targets for control.
Collapse
Affiliation(s)
- Daniel Plaugher
- Department of Toxicology and Cancer Biology, University of Kentucky
| | | |
Collapse
|
9
|
Giri SS, Tripathi AS, Erkekoğlu P, Zaki MEA. Molecular pathway of pancreatic cancer-associated neuropathic pain. J Biochem Mol Toxicol 2024; 38:e23638. [PMID: 38613466 DOI: 10.1002/jbt.23638] [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: 06/09/2023] [Revised: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 04/15/2024]
Abstract
The pancreas is a heterocrine gland that has both exocrine and endocrine parts. Most pancreatic cancer begins in the cells that line the ducts of the pancreas and is called pancreatic ductal adenocarcinoma (PDAC). PDAC is the most encountered pancreatic cancer type. One of the most important characteristic features of PDAC is neuropathy which is primarily due to perineural invasion (PNI). PNI develops tumor microenvironment which includes overexpression of fibroblasts cells, macrophages, as well as angiogenesis which can be responsible for neuropathy pain. In tumor microenvironment inactive fibroblasts are converted into an active form that is cancer-associated fibroblasts (CAFs). Neurotrophins they also increase the level of Substance P, calcitonin gene-related peptide which is also involved in pain. Matrix metalloproteases are the zinc-associated proteases enzymes which activates proinflammatory interleukin-1β into its activated form and are responsible for release and activation of Substance P which is responsible for neuropathic pain by transmitting pain signal via dorsal root ganglion. All the molecules and their role in being responsible for neuropathic pain are described below.
Collapse
Affiliation(s)
| | - Alok Shiomurti Tripathi
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, Uttar Pradesh, India
| | - Pınar Erkekoğlu
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Magdi E A Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad lbn Saud Islamic University, Riyadh, Saudi Arabia
| |
Collapse
|
10
|
Sun Y, Qin H, Zhang C, Xu J, Zhang T. Tetrastigma hemsleyanum (Sanyeqing) root extracts evoke S phase arrest while inhibiting the migration and invasion of human pancreatic cancer PANC-1 cells. BMC Complement Med Ther 2024; 24:133. [PMID: 38539165 PMCID: PMC10967071 DOI: 10.1186/s12906-024-04425-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/01/2024] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND Ethyl acetate extracts from Tetrastigma hemsleyanum (Sanyeqing) (EFT), a member of the Vitaceae plant family, have been shown to exhibit efficacy against a variety of cancers. In this light, our current study seeks to examine the mechanism of efficacy between EFT extracts and human pancreatic cancer PANC-1 cells. METHODS The chemical components of EFT were analyzed by gas chromatography-mass spectrometry. The cytotoxicity of EFT on PANC-1 cells was measured using an MTT assay. In order to investigate EFT induction of cell cycle arrest, changes in cell-cycle distribution were monitored by flow cytometry. Wound healing and transwell assays were employed to investigate whether migration and invasion of PANC-1 cells were inhibited by EFT. Relative protein expression was detected using Western blot. RESULTS GC-MS analysis of the chemical composition of EFT revealed that the majority of constituents were organic acids and their corresponding esters. EFT exhibits measurable cytotoxicity and inhibition of PANC-1 invasion. Growth inhibition was primarily attributed to downregulation of CDK2 which induces cell cycle arrest in the S-phase. Inhibition of metastasis is achieved through downregulation of mesenchymal-associated genes/activators, including ZEB1, N-cadherin, Vimentin, and Fibronectin. Meanwhile, the expression of E-cadherin was significantly increased by EFT treatment. Furthermore, downregulation of MMP-2 and MMP-9 were observed. CONCLUSION Treatment of PANC-1 with EFT demonstrated measurable cytotoxic effects. Furthermore, EFT evoked S phase arrest while inhibiting the migration and invasion of PANC-1 cells. Additionally, EFT inhibited the epithelial to mesenchymal transition and MMPs expression in PANC-1 cells. This study serves to confirm the strong therapeutic potential of EFT while identifying the mechanisms of action.
Collapse
Affiliation(s)
- Yifan Sun
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Binwen Road, Binjiang District, Hangzhou, Zhejiang Province, 310053, People's Republic of China
| | - Haiyan Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
- Nanjing Healthnice Pharmaceutical Technology Co., Ltd CN, Nanjing, 210031, People's Republic of China
| | - Chunchun Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Jian Xu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Binwen Road, Binjiang District, Hangzhou, Zhejiang Province, 310053, People's Republic of China
| | - Ting Zhang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Binwen Road, Binjiang District, Hangzhou, Zhejiang Province, 310053, People's Republic of China.
| |
Collapse
|
11
|
Murni W, Umar TP, Tandarto K, Simatupang A, Hasugian AR, Purwoko RY, Idaiani S, Stevanny B, Oktarina C, Jonlean R, Tango T, Kusuma KS, Sugiyono SP, Putra A. Efficacy and safety of medical cannabinoids in children with cerebral palsy: a systematic review. EINSTEIN-SAO PAULO 2023; 21:eRW0387. [PMID: 37991091 PMCID: PMC10691312 DOI: 10.31744/einstein_journal/2023rw0387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/19/2023] [Indexed: 11/23/2023] Open
Abstract
INTRODUCTION The increasing popularity of cannabinoids for treating numerous neurological disorders has been reported in various countries. Although it reduces tetrahydrocannabinol psychoactivity, it helps patients tolerate higher doses and complements the anti-spasmodic effects of tetrahydrocannabinol. One of the most important potential of cannabinoids are related to its potential to help children with cerebral palsy, a contributor of lifelong disability. Therefore, this systematic review aimed to assess the efficacy and safety of medical cannabinoids in children with cerebral palsy. METHODS This review adhered to The Preferred Reporting Items for Systematic Reviews and Meta-analysis 2020 guidelines. Seven databases, namely, Scopus, PubMed, EBSCO Host, ProQuest, Google Scholar, Semantic Scholar, and JSTOR, were used to identify relevant studies. Studies examining pediatric patients with cerebral palsy and reporting the efficacy and safety of medical cannabinoids through clinical trials, observational cross-sectional studies, or cohort designs were included. The outcomes of the studies included the efficacy of medical cannabinoids administered for spasticity, motor components, pain control, sleep difficulties, adverse effects, and seizure control. RESULTS Of 803 identified articles, only three met the inclusion criteria for data synthesis. One study exhibited a moderate risk-of-bias. A total of 133 respondents, mainly from Europe, were investigated. Overall effectiveness and safety were considered good. However, the results are inconsistent, especially regarding spasticity treatment variables. CONCLUSION The anti-spasticity, anti-inflammatory, and anti-seizure properties of cannabinoids might be beneficial for patients with cerebral palsy, although their effectiveness has not been widely studied. Further studies with larger sample sizes and various ethnicities are warranted. Prospero database registration: (www.crd.york.ac.uk/prospero) under ID CRD42022358383.
Collapse
Affiliation(s)
- Widya Murni
- Jakarta Anti-Aging Center ClinicJakartaIndonesia Jakarta Anti-Aging Center Clinic , Jakarta , Indonesia .
| | - Tungki Pratama Umar
- Faculty of MedicineSriwijaya UniversityPalembangIndonesia Faculty of Medicine , Sriwijaya University , Palembang , Indonesia .
| | - Kevin Tandarto
- Faculty of Medicine and Health SciencesAtma Jaya Catholic University of IndonesiaJakartaIndonesia Faculty of Medicine and Health Sciences , Atma Jaya Catholic University of Indonesia , Jakarta , Indonesia .
| | - Abraham Simatupang
- Department of Pharmacology and TherapyFaculty of MedicineUniversitas Kristen IndonesiaJakartaIndonesia Department of Pharmacology and Therapy , Faculty of Medicine , Universitas Kristen Indonesia , Jakarta , Indonesia .
| | - Armedy Ronny Hasugian
- Indonesia National Research and Innovation AgencyJakartaIndonesia Indonesia National Research and Innovation Agency , Jakarta , Indonesia .
| | - Reza Yuridian Purwoko
- Indonesia National Research and Innovation AgencyJakartaIndonesia Indonesia National Research and Innovation Agency , Jakarta , Indonesia .
| | - Sri Idaiani
- Indonesia National Research and Innovation AgencyJakartaIndonesia Indonesia National Research and Innovation Agency , Jakarta , Indonesia .
| | - Bella Stevanny
- Faculty of MedicineSriwijaya UniversityPalembangIndonesia Faculty of Medicine , Sriwijaya University , Palembang , Indonesia .
| | - Caroline Oktarina
- Faculty of MedicineUniversity of IndonesiaJakartaIndonesia Faculty of Medicine , University of Indonesia , Jakarta , Indonesia .
| | - Reganedgary Jonlean
- Faculty of MedicineUniversity of IndonesiaJakartaIndonesia Faculty of Medicine , University of Indonesia , Jakarta , Indonesia .
| | - Tamara Tango
- Faculty of MedicineUniversity of IndonesiaJakartaIndonesia Faculty of Medicine , University of Indonesia , Jakarta , Indonesia .
| | - Kevin Surya Kusuma
- Faculty of Medicine and Health SciencesAtma Jaya Catholic University of IndonesiaJakartaIndonesia Faculty of Medicine and Health Sciences , Atma Jaya Catholic University of Indonesia , Jakarta , Indonesia .
| | - Sagita Pratiwi Sugiyono
- Faculty of Medicine and Health SciencesAtma Jaya Catholic University of IndonesiaJakartaIndonesia Faculty of Medicine and Health Sciences , Atma Jaya Catholic University of Indonesia , Jakarta , Indonesia .
| | - Aditya Putra
- Faculty of Medicine and Health SciencesAtma Jaya Catholic University of IndonesiaJakartaIndonesia Faculty of Medicine and Health Sciences , Atma Jaya Catholic University of Indonesia , Jakarta , Indonesia .
| |
Collapse
|
12
|
Plaugher D, Murrugarra D. Phenotype Control techniques for Boolean gene regulatory networks. Bull Math Biol 2023; 85:89. [PMID: 37646851 PMCID: PMC10542862 DOI: 10.1007/s11538-023-01197-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/11/2023] [Indexed: 09/01/2023]
Abstract
Modeling cell signal transduction pathways via Boolean networks (BNs) has become an established method for analyzing intracellular communications over the last few decades. What's more, BNs provide a course-grained approach, not only to understanding molecular communications, but also for targeting pathway components that alter the long-term outcomes of the system. This has come to be known as phenotype control theory. In this review we study the interplay of various approaches for controlling gene regulatory networks such as: algebraic methods, control kernel, feedback vertex set, and stable motifs. The study will also include comparative discussion between the methods, using an established cancer model of T-Cell Large Granular Lymphocyte Leukemia. Further, we explore possible options for making the control search more efficient using reduction and modularity. Finally, we will include challenges presented such as the complexity and the availability of software for implementing each of these control techniques.
Collapse
Affiliation(s)
- Daniel Plaugher
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA.
| | - David Murrugarra
- Department of Mathematics, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
13
|
Plaugher D, Murrugarra D. Phenotype control techniques for Boolean gene regulatory networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.537158. [PMID: 37131770 PMCID: PMC10153207 DOI: 10.1101/2023.04.17.537158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Modeling cell signal transduction pathways via Boolean networks (BNs) has become an established method for analyzing intracellular communications over the last few decades. What’s more, BNs provide a course-grained approach, not only to understanding molecular communications, but also for targeting pathway components that alter the long-term outcomes of the system. This has come to be known as phenotype control theory . In this review we study the interplay of various approaches for controlling gene regulatory networks such as: algebraic methods, control kernel, feedback vertex set, and stable motifs. The study will also include comparative discussion between the methods, using an established cancer model of T-Cell Large Granular Lymphocyte (T-LGL) Leukemia. Further, we explore possible options for making the control search more efficient using reduction and modularity. Finally, we will include challenges presented such as the complexity and the availability of software for implementing each of these control techniques.
Collapse
Affiliation(s)
- Daniel Plaugher
- Department of Toxicology and Cancer Biology, University of Kentucky
| | | |
Collapse
|
14
|
Research advances and treatment perspectives of pancreatic adenosquamous carcinoma. Cell Oncol (Dordr) 2023; 46:1-15. [PMID: 36316580 DOI: 10.1007/s13402-022-00732-2] [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] [Accepted: 10/10/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND As a malignant tumor, pancreatic cancer has an extremely low overall 5-year survival rate. Pancreatic adenosquamous carcinoma (PASC), a rare pancreatic malignancy, owns clinical presentation similar to pancreatic ductal adenocarcinoma (PDAC), which is the most prevalent pancreatic cancer subtype. PASC is generally defined as a pancreatic tumor consisting mainly of adenocarcinoma tissue and squamous carcinoma tissue. Compared with PDAC, PASC has a higher metastatic potential and worse prognosis, and lacks of effective treatment options to date. However, the pathogenesis and treatment of PASC are not yet clear and are accompanied with difficulties. CONCLUSION The present paper systematically summarizes the possible pathogenesis, diagnosis methods, and further suggests potential new treatment directions through reviewing research results of PASC, including the clinical manifestations, pathological manifestation, the original hypothesis of squamous carcinoma and the potential regulatory mechanism. In short, the present paper provides a systematic review of the research progress and new ideas for the development mechanism and treatment of PASC.
Collapse
|
15
|
Szlasa W, Michel O, Sauer N, Novickij V, Lewandowski D, Kasperkiewicz P, Tarek M, Saczko J, Kulbacka J. Nanosecond pulsed electric field suppresses growth and reduces multi-drug resistance effect in pancreatic cancer. Sci Rep 2023; 13:351. [PMID: 36611083 PMCID: PMC9825384 DOI: 10.1038/s41598-023-27605-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
Nanosecond pulsed electric fields (nsPEF) have been shown to exert anticancer effects; however, little is known about the mechanisms triggered in cancer cells by nanosecond-length pulses, especially when low, sub-permeabilization voltage is used. In this study, three human pancreatic cancer cell lines were treated with nsPEF and molecular changes at the cellular level were analyzed. Further, we assessed the efficacy of paclitaxel chemotherapy following nsPEF treatment and correlated that with the changes in the expression of multi-drug resistance (MDR) proteins. Finally, we examined the influence of nsPEF on the adhesive properties of cancer cells as well as the formation and growth of pancreatic cancer spheroids. Cell line response differed with the application of a 200 ns, 100 pulses, 8 kV/cm, 10 kHz PEF treatment. PEF treatment led to (1) the release of microvesicles (MV) in EPP85-181RDB cells, (2) electropermeabilization in EPP85-181RNOV cells and (3) cell shrinkage in EPP85-181P cells. The release of MV's in EPP85-181RDB cells reduced the membrane content of P-gp and LRP, leading to a transient increase in vulnerability of the cells towards paclitaxel. In all cell lines we observed an initial reduction in size of the cancer spheroids after the nsPEF treatment. Cell line EPP85-181RNOV exhibited a permanent reduction in the spheroid size after nsPEF. We propose a mechanism in which the surface tension of the membrane, regulated by the organization of actin fibers, modulates the response of cancer cells towards nsPEF. When a membrane's surface tension remains low, we observed some cells form protrusions and release MVs containing MDR proteins. In contrast, when cell surface tension remains high, the cell membrane is being electroporated. The latter effect may be responsible for the reduced tumor growth following nsPEF treatment.
Collapse
Affiliation(s)
- Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland.
| | - Olga Michel
- grid.4495.c0000 0001 1090 049XDepartment of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland ,grid.8505.80000 0001 1010 5103Department of Cytobiochemistry, Faculty of Biotechnology, University of Wrocław, Wroclaw, Poland
| | - Natalia Sauer
- grid.4495.c0000 0001 1090 049XFaculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Vitalij Novickij
- grid.9424.b0000 0004 1937 1776Institute of High Magnetic Fields, Vilnius Gediminas Technical University, Vilnius, Lithuania ,grid.493509.2Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių 5, 08410 Vilnius, Lithuania
| | - Damian Lewandowski
- grid.8505.80000 0001 1010 5103Department of Animal Developmental Biology, Faculty of Biological Sciences, University of Wroclaw, Wroclaw, Poland
| | - Paulina Kasperkiewicz
- grid.7005.20000 0000 9805 3178Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Mounir Tarek
- grid.29172.3f0000 0001 2194 6418Université de Lorraine, CNRS, LPCT, 54000 Nancy, France
| | - Jolanta Saczko
- grid.4495.c0000 0001 1090 049XDepartment of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Julita Kulbacka
- grid.4495.c0000 0001 1090 049XDepartment of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland ,grid.493509.2Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių 5, 08410 Vilnius, Lithuania
| |
Collapse
|
16
|
Senturk ZN, Akdag I, Deniz B, Sayi-Yazgan A. Pancreatic cancer: Emerging field of regulatory B-cell-targeted immunotherapies. Front Immunol 2023; 14:1152551. [PMID: 37033931 PMCID: PMC10076755 DOI: 10.3389/fimmu.2023.1152551] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer, is characterized by a high mortality rate and poor prognosis. Current treatments for PDAC, are ineffective due to a prominent immunosuppressive PDAC tumor microenvironment (TME). Although B lymphocytes are highly infiltrated into PDAC, the importance of B lymphocytes in tumorigenesis is largely neglected. B cells play a dual role in the PDAC tumor microenvironment, acting as either anti-tumorigenic or pro-tumorigenic depending on where they are localized. Tumor-infiltrating B cells, which reside in ectopic lymph nodes, namely tertiary lymphoid structures (TLS), produce anti-tumor antibodies and present tumor antigens to T cells to contribute to cancer immunosurveillance. Alternatively, regulatory B cells (Bregs), dispersed inside the TME, contribute to the dampening of anti-tumor immune responses by secreting anti-inflammatory cytokines (IL-10 and IL-35), which promote tumor growth and metastasis. Determining the role of Bregs in the PDAC microenvironment is thus becoming increasingly attractive for developing novel immunotherapeutic approaches. In this minireview, we shed light on the emerging role of B cells in PDAC development and progression, with an emphasis on regulatory B cells (Bregs). Furthermore, we discussed the potential link of Bregs to immunotherapies in PDAC. These current findings will help us in understanding the full potential of B cells in immunotherapy.
Collapse
|
17
|
Plaugher D, Aguilar B, Murrugarra D. Uncovering potential interventions for pancreatic cancer patients via mathematical modeling. J Theor Biol 2022; 548:111197. [PMID: 35752283 DOI: 10.1016/j.jtbi.2022.111197] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/20/2022] [Accepted: 06/08/2022] [Indexed: 12/24/2022]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is widely known for its poor prognosis because it is often diagnosed when the cancer is in a later stage. We built a Boolean model to analyze the microenvironment of pancreatic cancer in order to better understand the interplay between pancreatic cancer, stellate cells, and their signaling cytokines. Specifically, we have used our model to study the impact of inducing four common mutations: KRAS, TP53, SMAD4, and CDKN2A. After implementing the various mutation combinations, we used our stochastic simulator to derive aggressiveness scores based on simulated attractor probabilities and long-term trajectory approximations. These aggression scores were then corroborated with clinical data. Moreover, we found sets of control targets that are effective among common mutations. These control sets contain nodes within both the pancreatic cancer cell and the pancreatic stellate cell, including PIP3, RAF, PIK3 and BAX in pancreatic cancer cell as well as ERK and PIK3 in the pancreatic stellate cell. Many of these nodes were found to be differentially expressed among pancreatic cancer patients in the TCGA database. Furthermore, literature suggests that many of these nodes can be targeted by drugs currently in circulation. The results herein help provide a proof of concept in the path towards personalized medicine through a means of mathematical systems biology. All data and code used for running simulations, statistical analysis, and plotting is available on a GitHub repository athttps://github.com/drplaugher/PCC_Mutations.
Collapse
Affiliation(s)
- Daniel Plaugher
- Department of Mathematics, University of Kentucky, Lexington, KY, USA.
| | | | - David Murrugarra
- Department of Mathematics, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
18
|
Monteiro MV, Ferreira LP, Rocha M, Gaspar VM, Mano JF. Advances in bioengineering pancreatic tumor-stroma physiomimetic Biomodels. Biomaterials 2022; 287:121653. [PMID: 35803021 DOI: 10.1016/j.biomaterials.2022.121653] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 01/18/2023]
Abstract
Pancreatic cancer exhibits a unique bioarchitecture and desmoplastic cancer-stoma interplay that governs disease progression, multi-resistance, and metastasis. Emulating the biological features and microenvironment heterogeneity of pancreatic cancer stroma in vitro is remarkably complex, yet highly desirable for advancing the discovery of innovative therapeutics. Diverse bioengineering approaches exploiting patient-derived organoids, cancer-on-a-chip platforms, and 3D bioprinted living constructs have been rapidly emerging in an endeavor to seamlessly recapitulate major tumor-stroma biodynamic interactions in a preclinical setting. Gathering on this, herein we showcase and discuss the most recent advances in bio-assembling pancreatic tumor-stroma models that mimic key disease hallmarks and its desmoplastic biosignature. A reverse engineering perspective of pancreatic tumor-stroma key elementary units is also provided and complemented by a detailed description of biodesign guidelines that are to be considered for improving 3D models physiomimetic features. This overview provides valuable examples and starting guidelines for researchers envisioning to engineer and characterize stroma-rich biomimetic tumor models. All in all, leveraging advanced bioengineering tools for capturing stromal heterogeneity and dynamics, opens new avenues toward generating more predictive and patient-personalized organotypic 3D in vitro platforms for screening transformative therapeutics targeting the tumor-stroma interplay.
Collapse
Affiliation(s)
- Maria V Monteiro
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Luís P Ferreira
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Marta Rocha
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Vítor M Gaspar
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
19
|
Richards KE, Xiao W, Hill R, on behalf of the USC Pancreas Research Team. Cancer-Associated Fibroblasts Confer Gemcitabine Resistance to Pancreatic Cancer Cells through PTEN-Targeting miRNAs in Exosomes. Cancers (Basel) 2022; 14:cancers14112812. [PMID: 35681792 PMCID: PMC9179363 DOI: 10.3390/cancers14112812] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/08/2022] [Accepted: 06/02/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Previous studies have shown that cancer associated fibroblasts exposed to chemotherapy release exosomes which promote chemoresistance in recipient cells. However, the molecular mechanism responsible for this has not been fully elucidated. In this study, we found that gemcitabine treatment caused fibroblasts to release exosome which contain PTEN-targeting miRNAs. These findings shed light on how fibroblasts exposed to chemotherapy promote tumor growth and drug resistance. Abstract Pancreatic ductal adenocarcinoma (PDAC) is currently the third leading cause of cancer-related death in the United States. Even though the poor prognosis of PDAC is often attributed to late diagnosis, patients with an early diagnosis who undergo tumor resection and adjuvant chemotherapy still show tumor recurrence, highlighting a need to develop therapies which can overcome chemoresistance. Chemoresistance has been linked to the high expression of microRNAs (miRs), such as miR-21, within tumor cells. Tumor cells can collect miRs through the uptake of miR-containing lipid extracellular vesicles called exosomes. These exosomes are secreted in high numbers from cancer-associated fibroblasts (CAFs) within the tumor microenvironment during gemcitabine treatment and can contribute to cell proliferation and chemoresistance. Here, we show a novel mechanism in which CAF-derived exosomes may promote proliferation and chemoresistance, in part, through suppression of the tumor suppressor PTEN. We identified five microRNAs: miR-21, miR-181a, miR-221, miR-222, and miR-92a, that significantly increased in number within the CAF exosomes secreted during gemcitabine treatment which target PTEN. Furthermore, we found that CAF exosomes suppressed PTEN expression in vitro and that treatment with the exosome inhibitor GW4869 blocked PTEN suppression in vivo. Collectively, these findings highlight a mechanism through which the PTEN expression loss, often seen in PDAC, may be attained and lend support to investigations into the use of exosome inhibitors as potential therapeutics to improve the effectiveness of chemotherapy.
Collapse
Affiliation(s)
- Katherine E. Richards
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 45556, USA;
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Weikun Xiao
- Lawrence J. Ellison Institute of Transformative Medicine, Los Angeles, CA 90064, USA;
| | - Reginald Hill
- Lawrence J. Ellison Institute of Transformative Medicine, Los Angeles, CA 90064, USA;
- Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
- Correspondence:
| | | |
Collapse
|
20
|
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an intractable cancer and a leading cause of cancer deaths worldwide. Over 90% of patients die within 1 year of diagnosis. Deaths from PDAC are increasing and it remains a cancer of substantial unmet need. A number of factors contribute to its poor prognosis: namely, late presentation, early metastases and limited systemic therapy options because of chemoresistance. A variety of research approaches underway are aimed at improving patient survival. Here, we review high-risk groups and efforts for early detection. We examine recent developments in the understanding of complex molecular and metabolic alterations which accompany PDAC. We explore artificial intelligence and biological targets for therapy and examine the role of tumour stroma and the immune microenvironment. We also review recent developments with respect to the PDAC microbiome. It is hoped that current research efforts will translate into earlier diagnosis, improvements in treatment and better outcomes for patients.
Collapse
Affiliation(s)
- Martyn C Stott
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| | - Lucy Oldfield
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| | - Jessica Hale
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| | - Eithne Costello
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| | - Christopher M Halloran
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| |
Collapse
|
21
|
Opitz FV, Haeberle L, Daum A, Esposito I. Tumor Microenvironment in Pancreatic Intraepithelial Neoplasia. Cancers (Basel) 2021; 13:cancers13246188. [PMID: 34944807 PMCID: PMC8699458 DOI: 10.3390/cancers13246188] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is a very aggressive neoplasm with a poor survival rate. This is mainly due to late detection, which substantially limits therapy options. A better understanding of the early phases of pancreatic carcinogenesis is fundamental for improving patient prognosis in the future. In this article, we focused on the tumor microenvironment (TME), which provides the biological niche for the development of PDAC from its most common precursor lesions, PanIN (pancreatic intraepithelial neoplasias). Abstract Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive tumors with a poor prognosis. A characteristic of PDAC is the formation of an immunosuppressive tumor microenvironment (TME) that facilitates bypassing of the immune surveillance. The TME consists of a desmoplastic stroma, largely composed of cancer-associated fibroblasts (CAFs), immunosuppressive immune cells, immunoregulatory soluble factors, neural network cells, and endothelial cells with complex interactions. PDAC develops from various precursor lesions such as pancreatic intraepithelial neoplasia (PanIN), intraductal papillary mucinous neoplasms (IPMN), mucinous cystic neoplasms (MCN), and possibly, atypical flat lesions (AFL). In this review, we focus on the composition of the TME in PanINs to reveal detailed insights into the complex restructuring of the TME at early time points in PDAC progression and to explore ways of modifying the TME to slow or even halt tumor progression.
Collapse
|
22
|
Ostios-Garcia L, Villamayor J, Garcia-Lorenzo E, Vinal D, Feliu J. Understanding the immune response and the current landscape of immunotherapy in pancreatic cancer. World J Gastroenterol 2021; 27:6775-6793. [PMID: 34790007 PMCID: PMC8567475 DOI: 10.3748/wjg.v27.i40.6775] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/28/2021] [Accepted: 09/19/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive tumor with high lethality. Even with surgery, radiotherapy, chemotherapy, and other locoregional or systemic therapies, the survival rates for PDAC are low and have not significantly changed in the past decades. The special characteristics of the PDAC's microenvironment and its complex immune escape mechanism need to be considered when designing novel therapeutic approaches in this disease. PDAC is characterized by chronic inflammation with a high rate of tumor-associated macrophages and myeloid-derived suppressor cells and a low rate of natural killer and effector T cells. The pancreatic microenvironment is a fibrotic, microvascularized stroma that isolates the tumor from systemic vascularization. Immunotherapy, a novel approach that has demonstrated effectiveness in certain solid tumors, has failed to show any practice-changing results in pancreatic cancer, with the exception of PDACs with mismatch repair deficiency and high tumor mutational burden, which show prolonged survival rates with immunotherapy. Currently, numerous clinical trials are attempting to assess the efficacy of immunotherapeutic strategies in PDAC, including immune checkpoint inhibitors, cancer vaccines, and adoptive cell transfer, alone or in combination with other immunotherapeutic agents, chemoradiotherapy, and other targeted therapies. A deep understanding of the immune response will help in the development of new therapeutic strategies leading to improved clinical outcomes for patients with PDAC.
Collapse
Affiliation(s)
- Lorena Ostios-Garcia
- Department of Oncology, La Paz University Hospital, IDIPAZ, CIBERONC, Cátedra UAM-AMGEN, Madrid 28046, Spain
| | - Julia Villamayor
- Department of Oncology, La Paz University Hospital, IDIPAZ, CIBERONC, Cátedra UAM-AMGEN, Madrid 28046, Spain
| | - Esther Garcia-Lorenzo
- Department of Oncology, La Paz University Hospital, IDIPAZ, CIBERONC, Cátedra UAM-AMGEN, Madrid 28046, Spain
| | - David Vinal
- Department of Oncology, La Paz University Hospital, IDIPAZ, CIBERONC, Cátedra UAM-AMGEN, Madrid 28046, Spain
| | - Jaime Feliu
- Department of Oncology, La Paz University Hospital, IDIPAZ, CIBERONC, Cátedra UAM-AMGEN, Madrid 28046, Spain
| |
Collapse
|
23
|
Gündel B, Liu X, Löhr M, Heuchel R. Pancreatic Ductal Adenocarcinoma: Preclinical in vitro and ex vivo Models. Front Cell Dev Biol 2021; 9:741162. [PMID: 34746135 PMCID: PMC8569794 DOI: 10.3389/fcell.2021.741162] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/04/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most overlooked cancers despite its dismal median survival time of 6 months. The biggest challenges in improving patient survival are late diagnosis due to lack of diagnostic markers, and limited treatment options due to almost complete therapy resistance. The past decades of research identified the dense stroma and the complex interplay/crosstalk between the cancer- and the different stromal cells as the main culprits for the slow progress in improving patient outcome. For better ex vivo simulation of this complex tumor microenvironment the models used in PDAC research likewise need to become more diverse. Depending on the focus of the investigation, several in vitro and in vivo models for PDAC have been established in the past years. Particularly, 3D cell culture such as spheroids and organoids have become more frequently used. This review aims to examine current PDAC in vitro models, their inherent limitations, and their successful implementations in research.
Collapse
Affiliation(s)
- Beate Gündel
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
| | - Xinyuan Liu
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
| | - Matthias Löhr
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
- Department of Upper GI, C1:77, Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Rainer Heuchel
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
24
|
Modeling the Pancreatic Cancer Microenvironment in Search of Control Targets. Bull Math Biol 2021; 83:115. [PMID: 34633559 DOI: 10.1007/s11538-021-00937-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022]
Abstract
Pancreatic ductal adenocarcinoma is among the leading causes of cancer-related deaths globally due to its extreme difficulty to detect and treat. Recently, research focus has shifted to analyzing the microenvironment of pancreatic cancer to better understand its key molecular mechanisms. This microenvironment can be represented with a multi-scale model consisting of pancreatic cancer cells (PCCs) and pancreatic stellate cells (PSCs), as well as cytokines and growth factors which are responsible for intercellular communication between the PCCs and PSCs. We have built a stochastic Boolean network (BN) model, validated by literature and clinical data, in which we probed for intervention strategies that force this gene regulatory network (GRN) from a diseased state to a healthy state. To do so, we implemented methods from phenotype control theory to determine a procedure for regulating specific genes within the microenvironment. We identified target genes and molecules, such that the application of their control drives the GRN to the desired state by suppression (or expression) and disruption of specific signaling pathways that may eventually lead to the eradication of the cancer cells. After applying well-studied control methods such as stable motifs, feedback vertex sets, and computational algebra, we discovered that each produces a different set of control targets that are not necessarily minimal nor unique. Yet, we were able to gain more insight about the performance of each process and the overlap of targets discovered. Nearly every control set contains cytokines, KRas, and HER2/neu, which suggests they are key players in the system's dynamics. To that end, this model can be used to produce further insight into the complex biological system of pancreatic cancer with hopes of finding new potential targets.
Collapse
|
25
|
Pijnappel EN, Wassenaar NPM, Gurney-Champion OJ, Klaassen R, van der Lee K, Pleunis-van Empel MCH, Richel DJ, Legdeur MC, Nederveen AJ, van Laarhoven HWM, Wilmink JW. Phase I/II Study of LDE225 in Combination with Gemcitabine and Nab-Paclitaxel in Patients with Metastatic Pancreatic Cancer. Cancers (Basel) 2021; 13:4869. [PMID: 34638351 PMCID: PMC8507646 DOI: 10.3390/cancers13194869] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Desmoplasia is a central feature of the tumor microenvironment in pancreatic ductal adenocarcinoma (PDAC). LDE225 is a pharmacological Hedgehog signaling pathway inhibitor and is thought to specifically target tumor stroma. We investigated the combined use of LDE225 and chemotherapy to treat PDAC patients. METHODS This was a multi-center, phase I/II study for patients with metastatic PDAC establishing the maximum tolerated dose of LDE225 co-administered with gemcitabine and nab-paclitaxel (phase I) and evaluating the efficacy and safety of the treatment combination after prior FOLFIRINOX treatment (phase II). Tumor microenvironment assessment was performed with quantitative MRI using intra-voxel incoherent motion diffusion weighted MRI (IVIM-DWI) and dynamic contrast-enhanced (DCE) MRI. RESULTS The MTD of LDE225 was 200 mg once daily co-administered with gemcitabine 1000 mg/m2 and nab-paclitaxel 125 mg/m2. In phase II, six therapy-related grade 4 adverse events (AE) and three grade 5 were observed. In 24 patients, the target lesion response was evaluable. Three patients had partial response (13%), 14 patients showed stable disease (58%), and 7 patients had progressive disease (29%). Median overall survival (OS) was 6 months (IQR 3.9-8.1). Blood plasma fraction (DCE) and diffusion coefficient (IVIM-DWI) significantly increased during treatment. Baseline perfusion fraction could predict OS (>222 days) with 80% sensitivity and 85% specificity. CONCLUSION LDE225 in combination with gemcitabine and nab-paclitaxel was well-tolerated in patients with metastatic PDAC and has promising efficacy after prior treatment with FOLFIRINOX. Quantitative MRI suggested that LDE225 causes increased tumor diffusion and works particularly well in patients with poor baseline tumor perfusion.
Collapse
Affiliation(s)
- Esther N. Pijnappel
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | - Nienke P. M. Wassenaar
- Cancer Center Amsterdam, Department of Radiology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (N.P.M.W.); (O.J.G.-C.); (A.J.N.)
| | - Oliver J. Gurney-Champion
- Cancer Center Amsterdam, Department of Radiology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (N.P.M.W.); (O.J.G.-C.); (A.J.N.)
| | - Remy Klaassen
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | - Koen van der Lee
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | | | - Dick J. Richel
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | - Marie C. Legdeur
- Department of Medical Oncology, Medisch Spectrum Twente, Twente, 7512 Enschede, The Netherlands; (M.C.H.P.-v.E.); (M.C.L.)
| | - Aart J. Nederveen
- Cancer Center Amsterdam, Department of Radiology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (N.P.M.W.); (O.J.G.-C.); (A.J.N.)
| | - Hanneke W. M. van Laarhoven
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | - Johanna W. Wilmink
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| |
Collapse
|
26
|
Romano R, Picca A, Eusebi LHU, Marzetti E, Calvani R, Moro L, Bucci C, Guerra F. Extracellular Vesicles and Pancreatic Cancer: Insights on the Roles of miRNA, lncRNA, and Protein Cargos in Cancer Progression. Cells 2021; 10:1361. [PMID: 34205944 PMCID: PMC8226820 DOI: 10.3390/cells10061361] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 01/18/2023] Open
Abstract
Pancreatic cancer (PC) is among the most devastating digestive tract cancers worldwide. This cancer is characterized by poor diagnostic detection, lack of therapy, and difficulty in predicting tumorigenesis progression. Although mutations of key oncogenes and oncosuppressor involved in tumor growth and in immunosurveillance escape are known, the underlying mechanisms that orchestrate PC initiation and progression are poorly understood or still under debate. In recent years, the attention of many researchers has been concentrated on the role of extracellular vesicles and of a particular subset of extracellular vesicles, known as exosomes. Literature data report that these nanovesicles are able to deliver their cargos to recipient cells playing key roles in the pathogenesis and progression of many pancreatic precancerous conditions. In this review, we have summarized and discussed principal cargos of extracellular vesicles characterized in PC, such as miRNAs, lncRNAs, and several proteins, to offer a systematic overview of their function in PC progression. The study of extracellular vesicles is allowing to understand that investigation of their secretion and analysis of their content might represent a new and potential diagnostic and prognostic tools for PC.
Collapse
Affiliation(s)
- Roberta Romano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy;
| | - Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (E.M.); (R.C.)
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institute and Stockholm University, 171 77 Stockholm, Sweden
| | - Leonardo Henry Umberto Eusebi
- Gastroenterology and Endoscopy Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Gastroenterology and Endoscopy Unit, Sant’Orsola University Hospital, 40138 Bologna, Italy
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (E.M.); (R.C.)
- Institute of Internal Medicine and Geriatrics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (E.M.); (R.C.)
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institute and Stockholm University, 171 77 Stockholm, Sweden
| | - Loredana Moro
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA; or
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY 10016, USA
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, 70126 Bari, Italy
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy;
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy;
| |
Collapse
|
27
|
Hama M, Ishima Y, Chuang VTG, Ando H, Shimizu T, Ishida T. Evidence for Delivery of Abraxane via a Denatured-Albumin Transport System. ACS APPLIED MATERIALS & INTERFACES 2021; 13:19736-19744. [PMID: 33881292 DOI: 10.1021/acsami.1c03065] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Abraxane, an albumin-bound paclitaxel nanoparticle formulation, is superior to conventional paclitaxel preparations because it has better efficacy against unresectable pancreatic cancer. Previous reports suggest that this better efficacy of Abraxane than conventional paclitaxel preparation is probably due to its transport through Gp60, an albumin receptor on the surface of vascular endothelial cells. The increased tumor accumulation of Abraxane is also caused by the secreted protein acid and rich in cysteine in the tumor stroma. However, the uptake mechanism of Abraxane remains poorly understood. In this study, we demonstrated that the delivery of Abraxane occurred via different receptor pathways from that of endogenous albumin. Our results showed that the uptake of endogenous albumin was inhibited by a Gp60 pathway inhibitor in the process of endocytosis through endothelial cells or tumor cells. In contrast, the uptake of Abraxane-derived HSA was less affected by the Gp60 pathway inhibitor but significantly reduced by denatured albumin receptor inhibitors. In conclusion, these data indicate that Abraxane-derived HSA was taken up into endothelial cells or tumor cells by a mechanism different from normal endogenous albumin. These new data on distinct cellular transport pathways of denatured albumin via gp family proteins different from those of innate albumin shed light on the mechanisms of tumor delivery and antitumor activity of Abraxane and provide new scientific rationale for the development of a novel albumin drug delivery strategy via a denatured albumin receptor.
Collapse
Affiliation(s)
- Maichi Hama
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Yu Ishima
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Victor Tuan Giam Chuang
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Western Australia 6102, Australia
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| |
Collapse
|
28
|
Rai ZL, Feakins R, Pallett LJ, Manas D, Davidson BR. Irreversible Electroporation (IRE) in Locally Advanced Pancreatic Cancer: A Review of Current Clinical Outcomes, Mechanism of Action and Opportunities for Synergistic Therapy. J Clin Med 2021; 10:1609. [PMID: 33920118 PMCID: PMC8068938 DOI: 10.3390/jcm10081609] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/04/2021] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
Locally advanced pancreatic cancer (LAPC) accounts for 30% of patients with pancreatic cancer. Irreversible electroporation (IRE) is a novel cancer treatment that may improve survival and quality of life in LAPC. This narrative review will provide a perspective on the clinical experience of pancreas IRE therapy, explore the evidence for the mode of action, assess treatment complications, and propose strategies for augmenting IRE response. A systematic search was performed using PubMed regarding the clinical use and safety profile of IRE on pancreatic cancer, post-IRE sequential histological changes, associated immune response, and synergistic therapies. Animal data demonstrate that IRE induces both apoptosis and necrosis followed by fibrosis. Major complications may result from IRE; procedure related mortality is up to 2%, with an average morbidity as high as 36%. Nevertheless, prospective and retrospective studies suggest that IRE treatment may increase median overall survival of LAPC to as much as 30 months and provide preliminary data justifying the well-designed trials currently underway, comparing IRE to the standard of care treatment. The mechanism of action of IRE remains unknown, and there is a lack of data on treatment variables and efficiency in humans. There is emerging data suggesting that IRE can be augmented with synergistic therapies such as immunotherapy.
Collapse
Affiliation(s)
- Zainab L. Rai
- Centre of Surgical Innovation, Organ Regeneration and Transplantation, University College London (UCL), London NW3 2QG, UK;
- Wellcome/EPSRC Center for Interventional and Surgical Sciences (WEISS), London W1W 7TY, UK
- Royal Free NHS Foundation Trust, London NW3 2QG, UK;
| | - Roger Feakins
- Royal Free NHS Foundation Trust, London NW3 2QG, UK;
| | - Laura J. Pallett
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London WC1E 6BT, UK;
| | - Derek Manas
- Newcastle Upon Tyne NHS Foundation Trust, Newcastle-Upon-Tyne NE7 7DN, UK;
| | - Brian R. Davidson
- Centre of Surgical Innovation, Organ Regeneration and Transplantation, University College London (UCL), London NW3 2QG, UK;
- Royal Free NHS Foundation Trust, London NW3 2QG, UK;
| |
Collapse
|
29
|
Dailey KM, Allgood JE, Johnson PR, Ostlie MA, Schaner KC, Brooks BD, Brooks AE. The next frontier of oncotherapy: accomplishing clinical translation of oncolytic bacteria through genetic engineering. Future Microbiol 2021; 16:341-368. [PMID: 33754804 DOI: 10.2217/fmb-2020-0245] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The development of a 'smart' drug capable of distinguishing tumor from host cells has been sought for centuries, but the microenvironment of solid tumors continues to confound therapeutics. Solid tumors present several challenges for current oncotherapeutics, including aberrant vascularization, hypoxia, necrosis, abnormally high pH and local immune suppression. While traditional chemotherapeutics are limited by such an environment, oncolytic microbes are drawn to it - having an innate ability to selectively infect, colonize and eradicate solid tumors. Development of an oncolytic species would represent a shift in the cancer therapeutic paradigm, with ramifications reaching from the medical into the socio-economic. Modern genetic engineering techniques could be implemented to customize 'Frankenstein' bacteria with advantageous characteristics from several species.
Collapse
Affiliation(s)
- Kaitlin M Dailey
- Cellular & Molecular Biology Program, North Dakota State University, Fargo, ND 58103, USA.,Pharmaceutical Sciences Department, North Dakota State University, Fargo, ND 58103, USA
| | - JuliAnne E Allgood
- Department of Neuroscience, University of Wyoming, Laramie, WY 82071, USA
| | - Paige R Johnson
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, ND 58103, USA
| | - Mackenzie A Ostlie
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, ND 58103, USA
| | - Kambri C Schaner
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, ND 58103, USA
| | | | - Amanda E Brooks
- Cellular & Molecular Biology Program, North Dakota State University, Fargo, ND 58103, USA.,Pharmaceutical Sciences Department, North Dakota State University, Fargo, ND 58103, USA.,Office of Research & Scholarly Activity. Rocky Vista University, Ivins, UT 84738, USA
| |
Collapse
|
30
|
Role of targeted immunotherapy for pancreatic ductal adenocarcinoma (PDAC) treatment: An overview. Int Immunopharmacol 2021; 95:107508. [PMID: 33725635 DOI: 10.1016/j.intimp.2021.107508] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/18/2021] [Accepted: 02/12/2021] [Indexed: 12/15/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest solid tumors with a high mortality rate and poor survival rate. Depending on the tumor stage, PDAC is either treated by resection surgery, chemotherapies, or radiotherapies. Various chemotherapeutic agents have been used to treat PDAC, alone or in combination. Despite the combinations, chemotherapy exhibits many side-effects leading to an increase in the toxicity profile amongst the PDAC patients. Additionally, these standard chemotherapeutic agents have only a modest impact on patient survival due to their limited efficacy. PDAC was previously considered as an immunologically silent malignancy, but recent findings have demonstrated that effective immune-mediated tumor cell death can be used for its treatment. PDAC is characterized by an immunosuppressive tumor microenvironment accompanied by the major expression of myeloid-derived suppressor cells (MDSC) and M2 tumor-associated macrophages. In contrast, the expression of CD8+ T cells is significantly low. Additionally, infiltration of mast cells in PDAC correlates with the poor prognosis. Immunotherapeutic agents target the immunity mediators and empower them to suppress the tumor and effectively treat PDAC. Different targets are studied and exploited to induce an antitumor immune response in PDAC patients. In recent times, site-specific delivery of immunotherapeutics also gained attention among researchers to effectively treat PDAC. In the present review, existing immunotherapies for PDAC treatment along with their limitations are addressed in detail. The review also includes the pathophysiology, traditional strategies and significance of targeted immunotherapies to combat PDAC effectively. Separately, the identification of ideal targets for the targeted therapy of PDAC is also reviewed exhaustively. Additionally, the review also addresses the applications of targeted immunotherapeutics like checkpoint inhibitors, adoptive T-cell therapy etc.
Collapse
|
31
|
Holbrook MC, Goad DW, Grdzelishvili VZ. Expanding the Spectrum of Pancreatic Cancers Responsive to Vesicular Stomatitis Virus-Based Oncolytic Virotherapy: Challenges and Solutions. Cancers (Basel) 2021; 13:1171. [PMID: 33803211 PMCID: PMC7963195 DOI: 10.3390/cancers13051171] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating malignancy with poor prognosis and a dismal survival rate, expected to become the second leading cause of cancer-related deaths in the United States. Oncolytic virus (OV) is an anticancer approach that utilizes replication-competent viruses to preferentially infect and kill tumor cells. Vesicular stomatitis virus (VSV), one such OV, is already in several phase I clinical trials against different malignancies. VSV-based recombinant viruses are effective OVs against a majority of tested PDAC cell lines. However, some PDAC cell lines are resistant to VSV. Upregulated type I IFN signaling and constitutive expression of a subset of interferon-simulated genes (ISGs) play a major role in such resistance, while other mechanisms, such as inefficient viral attachment and resistance to VSV-mediated apoptosis, also play a role in some PDACs. Several alternative approaches have been shown to break the resistance of PDACs to VSV without compromising VSV oncoselectivity, including (i) combinations of VSV with JAK1/2 inhibitors (such as ruxolitinib); (ii) triple combinations of VSV with ruxolitinib and polycations improving both VSV replication and attachment; (iii) combinations of VSV with chemotherapeutic drugs (such as paclitaxel) arresting cells in the G2/M phase; (iv) arming VSV with p53 transgenes; (v) directed evolution approach producing more effective OVs. The latter study demonstrated impressive long-term genomic stability of complex VSV recombinants encoding large transgenes, supporting further clinical development of VSV as safe therapeutics for PDAC.
Collapse
Affiliation(s)
| | | | - Valery Z. Grdzelishvili
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; (M.C.H.); (D.W.G.)
| |
Collapse
|
32
|
Zhang C, Dang D, Liu C, Wang Y, Cong X. Identification of tumor mutation burden-related hub genes and the underlying mechanism in melanoma. J Cancer 2021; 12:2440-2449. [PMID: 33758620 PMCID: PMC7974884 DOI: 10.7150/jca.53697] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Tumor mutation burden (TMB) has emerged as an important predictive factor for drug resistance in cancers; however, the specific mechanism underlying TMB function in melanoma remains elusive. Methods: Data on somatic mutations, RNA sequencing (RNA-seq), miRNA sequencing (miRNA-seq), and clinical characteristics for 472 melanoma patients were extracted from the TCGA cohort. RNA-seq data of melanoma cell lines were obtained from the Cancer Cell Line Encyclopedia, and sensitivity of cell lines to therapeutic agents is available in the Cancer Therapeutics Response Portal. TMB was calculated based on somatic mutation data. Differentially expressed gene analysis, weighted gene co-expression network analysis, protein-protein interaction networks, Minimal Common Oncology Data Elements, and survival analysis were leveraged to determine TMB-related hub genes. Competing endogenous RNA (ceRNA) networks were constructed to explore the molecular mechanisms underlying hub gene function. The influence of key genes on drug sensitivity was analyzed to investigate their clinical significance. Results: Elevated TMB levels were significantly correlated with improved survival outcomes. In addition, six tumor-infiltrating immune cells, including naive B cells, regulatory T cells, memory resting CD4 T cells, memory B cells, activated mast cells, and resting NK cells, were significantly overexpressed in the low-TMB group relative to the high-TMB group. Furthermore, we identified FLNC, NEXN, and TNNT3 as TMB-related hub genes, and constructed their ceRNA networks, including five miRNAs (has-miR-590-3p, has-miR-374b-5p, has-miR-3127-5p, has-miR-1913, and has-miR-1291) and 31 lncRNAs (FAM66C, MIAT, NR2F2AS1, etc.). Finally, we observed that TMB-related genes were associated with distinct therapeutic responses to AKT/mTOR pathway inhibitors. Conclusions: We identified three TMB-associated key genes, established their ceRNA networks, and investigated their influence on therapeutic responses, which could provide insights into future precision medicine.
Collapse
Affiliation(s)
- Chuan Zhang
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China
- Department of Pediatric Surgery, the First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Dan Dang
- Department of Neonatology, the First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Chenlu Liu
- Department of Tissue Bank, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China
| | - Yuqian Wang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China
| | - Xianling Cong
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China
| |
Collapse
|
33
|
Gaida MM. [The ambiguous role of the inflammatory micromilieu in solid tumors]. DER PATHOLOGE 2021; 41:118-123. [PMID: 33104890 DOI: 10.1007/s00292-020-00837-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Besides host defense against infections, the main function of the immune system is to eliminate tumor cells. Therefore, nearly, all solid tumors have a heterogeneous fibro-inflammatory microenvironment, which consists of myofibroblastic cells, extracellular matrix components, and infiltrates from various types of immune cell. In particular, pancreatic ductal adenocarcinoma is a prototype of a tumor with a pronounced inflammatory microenvironment, in which the majority of the tumor mass consists of nonneoplastic stromal and immune cells. Our own data and data from the literature indicate a protective role of tumor-infiltrating T cells for the host. On the other hand, we were able to show that a defined T cell subpopulation paradoxically promotes the progression of the tumor. Our investigations now focus on these cells, known as "Th17," in the tumor microenvironment. OBJECTIVES To elucidate the mechanisms of the infiltrated immune cells and their mediators in the tumor microenvironment. MATERIALS AND METHODS Human pancreatic cancer tissue was used for (immune) histological staining and morphometric analysis and the results were correlated with clinical parameters and with diffusion-weighted magnetic resonance imaging images. The molecular mechanisms were analyzed in cell culture approaches using human tumor cells and human immune cells. With molecular biological methods and functional assays cell growth, invasion and colony formation were assessed. The in vivo correlation of the results and functional interventions were tested in murine and avian (xenograft) models. RESULTS AND CONCLUSION Tumor-infiltrating immune cells of type Th17 and their mediators promoted the progression of the tumor depending on density, activation status, and cytokine profile. On molecular level, we identified a Th17-mediated increase of tumor cell migration and invasion, an increased neoangiogenesis, as well as a reorganization of the tumor stroma and microarchitecture. The data show that the progression of pancreatic cancer, depends on the status of activation and the cytokine profile of the infiltrated T cells.
Collapse
Affiliation(s)
- Matthias M Gaida
- Institut für Pathologie, Universitätsmedizin der Johannes Gutenberg-Universität Mainz, Langenbeckstr. 1, 55131, Mainz, Deutschland.
| |
Collapse
|
34
|
Khomiak A, Brunner M, Kordes M, Lindblad S, Miksch RC, Öhlund D, Regel I. Recent Discoveries of Diagnostic, Prognostic and Predictive Biomarkers for Pancreatic Cancer. Cancers (Basel) 2020; 12:E3234. [PMID: 33147766 PMCID: PMC7692691 DOI: 10.3390/cancers12113234] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with a dismal prognosis that is frequently diagnosed at an advanced stage. Although less common than other malignant diseases, it currently ranks as the fourth most common cause of cancer-related death in the European Union with a five-year survival rate of below 9%. Surgical resection, followed by adjuvant chemotherapy, remains the only potentially curative treatment but only a minority of patients is diagnosed with locally resectable, non-metastatic disease. Patients with advanced disease are treated with chemotherapy but high rates of treatment resistance and unfavorable side-effect profiles of some of the used regimens remain major challenges. Biomarkers reflect pathophysiological or physiological processes linked to a disease and can be used as diagnostic, prognostic and predictive tools. Thus, accurate biomarkers can allow for better patient stratification and guide therapy choices. Currently, the only broadly used biomarker for PDAC, CA 19-9, has multiple limitations and the need for novel biomarkers is urgent. In this review, we highlight the current situation, recent discoveries and developments in the field of biomarkers of PDAC and their potential clinical applications.
Collapse
Affiliation(s)
- Andrii Khomiak
- Shalimov National Institute of Surgery and Transplantology, 03058 Kyiv, Ukraine;
| | - Marius Brunner
- Department of Gastroenterology, Endocrinology and Gastrointestinal Oncology, University Medical Center, 37075 Goettingen, Germany;
| | - Maximilian Kordes
- Department of Upper Abdominal Diseases, Karolinska University Hospital, 14186 Stockholm, Sweden;
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 17177 Stockholm, Sweden
| | - Stina Lindblad
- Department of Radiation Sciences, Sweden and Wallenberg Centre for Molecular Medicine, Umeå University, 90187 Umeå, Sweden;
| | - Rainer Christoph Miksch
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, 81377 Munich, Germany;
| | - Daniel Öhlund
- Department of Radiation Sciences, Sweden and Wallenberg Centre for Molecular Medicine, Umeå University, 90187 Umeå, Sweden;
| | - Ivonne Regel
- Department of Medicine II, University Hospital, LMU Munich, 81377 Munich, Germany
| |
Collapse
|
35
|
Qorri B, Harless W, Szewczuk MR. Novel Molecular Mechanism of Aspirin and Celecoxib Targeting Mammalian Neuraminidase-1 Impedes Epidermal Growth Factor Receptor Signaling Axis and Induces Apoptosis in Pancreatic Cancer Cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:4149-4167. [PMID: 33116404 PMCID: PMC7550724 DOI: 10.2147/dddt.s264122] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/27/2020] [Indexed: 12/11/2022]
Abstract
Background Aspirin (acetylsalicylic acid) and celecoxib have been used as potential anti-cancer therapies. Aspirin exerts its therapeutic effect in both cyclooxygenase (COX)-dependent and -independent pathways to reduce tumor growth and disable tumorigenesis. Celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor, reduces factors that cause inflammation and pain. The question is whether aspirin and celecoxib have other molecular targets of equal or more therapeutic efficacy with significant anti-cancer preventive benefits. Aim Here, we propose that aspirin and celecoxib exert their anti-cancer effects by targeting and inhibiting mammalian neuraminidase-1 (Neu-1). Neu-1 has been reported to regulate the activation of several receptor tyrosine kinases (RTKs) and TOLL-like receptors and their downstream signaling pathways. Neu-1 in complex with matrix metalloproteinase-9 (MMP-9) and G protein-coupled receptors (GPCRs) has been reported to be tethered to RTKs at the ectodomain. Materials and Methods The WST-1 cell viability assay, Caspase 3/7 assay, and Annexin V assay were used to evaluate the cell viability and detect apoptotic and necrotic cells following treatment in MiaPaCa-2, PANC-1 and the gemcitabine-resistant PANC-1 variant (PANC-1 GemR) cells. Microscopic imaging, lectin cytochemistry, and flow cytometry were used to detect levels of α-2,3 sialic acid. Epidermal growth factor (EGF)-stimulated live cell sialidase assays and neuraminidase assays were used to detect Neu-1 activity. Immunocytochemistry was used to detect levels of EGFR and phosphorylated EGFR (pEGFR) following treatment. Results For the first time, aspirin and celecoxib were shown to significantly inhibit Neu-1 sialidase activity in a dose- and time-dependent manner following stimulation with EGF. Aspirin blocked Neu-1 desialylation of α-2,3-sialic acid expression following 30 min stimulation with EGF. Aspirin and celecoxib significantly and dose-dependently inhibited isolated neuraminidase (Clostridium perfringens) activity on fluorogenic substrate 2ʹ-(4-methylumbelliferyl)-α-D-N-acetylneuraminic acid (4-MUNANA). Aspirin inhibited phosphorylation of the EGFR in EGF-stimulated cells. Aspirin dose- and time-dependently induced CellEvent caspase-3/7+ cells as well as apoptosis and necrosis on PANC-1 cells. Conclusion These findings signify a novel multimodality mechanism(s) of action for aspirin and celecoxib, specifically targeting and inhibiting Neu-1 activity, regulating EGF-induced growth receptor activation and inducing apoptosis and necrosis in a dose- and time-dependent manner. Repurposing aspirin and celecoxib as anti-cancer agents may also upend other critical targets involved in multistage tumorigenesis regulated by mammalian neuraminidase-1. Significance These findings may be the missing link connecting the anti-cancer efficacy of NSAIDs to the role of glycosylation in inflammation and tumorigenesis.
Collapse
Affiliation(s)
- Bessi Qorri
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | | | - Myron R Szewczuk
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| |
Collapse
|
36
|
Turkes F, Mencel J, Starling N. Targeting the immune milieu in gastrointestinal cancers. J Gastroenterol 2020; 55:909-926. [PMID: 32748171 PMCID: PMC7519898 DOI: 10.1007/s00535-020-01710-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023]
Abstract
Gastrointestinal (GI) cancers are among the most common and lethal solid tumors worldwide. Unlike in malignancies such as lung, renal and skin cancers, the activity of immunotherapeutic agents in GI cancers has, on the whole, been much less remarkable and do not apply to the majority. Furthermore, while incremental progress has been made and approvals for use of immune checkpoint inhibitors (ICIs) in specific subsets of patients with GI cancers are coming through, in a population of 'all-comers', it is frequently unclear as to who may benefit most due to the relative lack of reliable predictive biomarkers. For most patients with newly diagnosed advanced or metastatic GI cancer, the mainstay of treatment still involves chemotherapy and/or a targeted agent however, beyond the second-line this paradigm confers minimal patient benefit. Thus, current research efforts are concentrating on broadening the applicability of ICIs in GI cancers by combining them with agents designed to beneficially remodel the tumor microenvironment (TME) for more effective anti-cancer immunity with intention of improving patient outcomes. This review will discuss the currently approved ICIs available for the treatment of GI cancers, the strategies underway focusing on combining ICIs with agents that target the TME and touch on recent progress toward identification of predictors of sensitivity to immune checkpoint blockade in GI cancers.
Collapse
Affiliation(s)
- Fiona Turkes
- Department of Medicine, Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Justin Mencel
- Department of Medicine, Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Naureen Starling
- Department of Medicine, Royal Marsden Hospital NHS Foundation Trust, London, UK.
| |
Collapse
|
37
|
Van Audenaerde JR, Marcq E, von Scheidt B, Davey AS, Oliver AJ, De Waele J, Quatannens D, Van Loenhout J, Pauwels P, Roeyen G, Lardon F, Slaney CY, Peeters M, Kershaw MH, Darcy PK, Smits EL. Novel combination immunotherapy for pancreatic cancer: potent anti-tumor effects with CD40 agonist and interleukin-15 treatment. Clin Transl Immunology 2020; 9:e1165. [PMID: 32821382 PMCID: PMC7428816 DOI: 10.1002/cti2.1165] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/08/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
Objectives With the poorest 5‐year survival of all cancers, improving treatment for pancreatic cancer is one of the biggest challenges in cancer research. We sought to explore the potential of combining both priming and activation of the immune system. To achieve this, we combined a CD40 agonist with interleukin‐15 and tested its potential in pancreatic cancer. Methods Response to this combination regimen was assessed in pancreatic ductal adenocarcinoma mouse models, and a thorough analysis of the tumor microenvironment was performed. Results We demonstrated profound reduction in tumor growth and increased survival of mice with the majority of mice being cured when both agents were combined, including an unprecedented 8‐fold dose reduction of CD40 agonist without losing any efficacy. RNAseq analysis showed involvement of natural killer (NK) cell‐ and T‐cell‐mediated anti‐tumor responses and the importance of antigen‐presenting cell pathways. This combination resulted in enhanced infiltration of tumors by both T cells and NK cells, as well as a striking increase in the ratio of CD8+ T cells over Tregs. We also observed a significant increase in numbers of dendritic cells (DCs) in tumor‐draining lymph nodes, particularly CD103+ DCs with cross‐presentation potential. A critical role for CD8+ T cells and involvement of NK cells in the anti‐tumor effect was highlighted. Importantly, strong immune memory was established, with an increase in memory CD8+ T cells only when both interleukin‐15 and the CD40 agonist were combined. Conclusion These novel preclinical data support initiation of a first‐in‐human clinical trial with this combination immunotherapy strategy in pancreatic cancer.
Collapse
Affiliation(s)
- Jonas Rm Van Audenaerde
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium.,Cancer Immunotherapy and Immune Innovation Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia
| | - Elly Marcq
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium
| | - Bianca von Scheidt
- Cancer Immunotherapy and Immune Innovation Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia
| | - Ashleigh S Davey
- Cancer Immunotherapy and Immune Innovation Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia
| | - Amanda J Oliver
- Cancer Immunotherapy and Immune Innovation Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia
| | - Jorrit De Waele
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium
| | - Delphine Quatannens
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium
| | - Jinthe Van Loenhout
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium
| | - Patrick Pauwels
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium.,Department of Pathology Antwerp University Hospital Edegem Belgium
| | - Geert Roeyen
- Department of Hepatobiliary, Endocrine and Transplantation Surgery Antwerp University Hospital Edegem Belgium
| | - Filip Lardon
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium
| | - Clare Y Slaney
- Cancer Immunotherapy and Immune Innovation Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Marc Peeters
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium.,Department of Oncology and Multidisciplinary Oncological Centre Antwerp Antwerp University Hospital Edegem Belgium
| | - Michael H Kershaw
- Cancer Immunotherapy and Immune Innovation Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Phillip K Darcy
- Cancer Immunotherapy and Immune Innovation Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Evelien Ljm Smits
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium.,Center for Cell Therapy and Regenerative Medicine Antwerp University Hospital Edegem Belgium
| |
Collapse
|
38
|
Lu J, Wei S, Lou J, Yin S, Zhou L, Zhang W, Zheng S. Systematic Analysis of Alternative Splicing Landscape in Pancreatic Adenocarcinoma Reveals Regulatory Network Associated with Tumorigenesis and Immune Response. Med Sci Monit 2020; 26:e925733. [PMID: 32706768 PMCID: PMC7709468 DOI: 10.12659/msm.925733] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive gastrointestinal tumors and has an extremely high mortality rate. Recent studies indicate that alternative splicing (AS), a common post-transcriptional process, has important roles in tumor biological behaviors and may provide novel immunotherapeutic targets. This study systematically analyzes AS profiles in PDAC and reveals their potential regulatory effects on cancer immune response. MATERIAL AND METHODS AS event, RNA sequencing, and splicing factor (SF) data were extracted from SpliceSeq, The Cancer Genome Atlas, and SpliceAid2, respectively. Overall survival (OS)-associated AS events and SFs were identified with univariate analysis. The LASSO method and multivariate Cox regression analysis were used to construct predictive signatures for the prediction of patient prognosis. The proportions of immune cells within PDAC samples were evaluated using the CIBERSORT algorithm. The correlations among AS events, SFs, and immune cell proportions were calculated using Spearman correlation analysis. Consensus clustering and immune classification were performed on the PDAC cohort. RESULTS A total of 4812 OS-related AS events from 3341 parent genes were identified, and 8 AS-based predictive models were constructed for PDAC. An OS-related SF-AS regulatory network was constructed. The AS events regulated by ELAVL4 exhibited strong correlations with CD8 T cells and regulatory T cells. In addition, AS-based clusters demonstrated distinct OS outcomes and immune features. CONCLUSIONS AS-based predictive models with high accuracy were constructed to facilitate prognosis prediction and treatment of PDAC. An SF-AS regulatory network was constructed, revealing the potential relationships among SF, AS, and immune response.
Collapse
Affiliation(s)
- Jiahua Lu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, Zhejiang, China (mainland).,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, China (mainland).,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang, China (mainland)
| | - Shenyu Wei
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China (mainland)
| | - Jianying Lou
- Department of Hepato-Pancreato-Biliary Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Shengyong Yin
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, Zhejiang, China (mainland).,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, China (mainland).,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang, China (mainland)
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, Zhejiang, China (mainland).,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, China (mainland).,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang, China (mainland)
| | - Wu Zhang
- Shulan (Hangzhou) Hospital, Hangzhou, Zhejiang, China (mainland).,School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, Zhejiang, China (mainland).,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang, China (mainland).,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
39
|
Awaji M, Saxena S, Wu L, Prajapati DR, Purohit A, Varney ML, Kumar S, Rachagani S, Ly QP, Jain M, Batra SK, Singh RK. CXCR2 signaling promotes secretory cancer-associated fibroblasts in pancreatic ductal adenocarcinoma. FASEB J 2020; 34:9405-9418. [PMID: 32453916 PMCID: PMC7501205 DOI: 10.1096/fj.201902990r] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/21/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most challenging malignancies. Desmoplasia and tumor-supporting inflammation are hallmarks of PDAC. The tumor microenvironment contributes significantly to tumor progression and spread. Cancer-associated fibroblasts (CAFs) facilitate therapy resistance and metastasis. Recent reports emphasized the concurrence of multiple subtypes of CAFs with diverse roles, fibrogenic, and secretory. C-X-C motif chemokine receptor 2 (CXCR2) is a chemokine receptor known for its role during inflammation and its adverse role in PDAC. Oncogenic Kras upregulates CXCR2 and its ligands and, thus, contribute to tumor proliferation and immunosuppression. CXCR2 deletion in a PDAC syngeneic mouse model produced increased fibrosis revealing a potential undescribed role of CXCR2 in CAFs. In this study, we demonstrate that the oncogenic Kras-CXCR2 axis regulates the CAFs function in PDAC and contributes to CAFs heterogeneity. We observed that oncogenic Kras and CXCR2 signaling alter CAFs, producing a secretory CAF phenotype with low fibrogenic features; and increased secretion of pro-tumor cytokines and CXCR2 ligands, utilizing the NF-κB activity. Finally, using syngeneic mouse models, we demonstrate that oncogenic Kras is associated with secretory CAFs and that CXCR2 inhibition promotes activation of fibrotic cells (myofibroblasts) and impact tumors in a mutation-dependent manner.
Collapse
MESH Headings
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cancer-Associated Fibroblasts/metabolism
- Cancer-Associated Fibroblasts/pathology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cell Proliferation
- Gene Expression Regulation, Neoplastic
- Mice
- Mice, Knockout
- Mutation
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/metabolism
- Signal Transduction
- Tumor Cells, Cultured
- Tumor Microenvironment
- Pancreatic Neoplasms
Collapse
Affiliation(s)
- Mohammad Awaji
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha NE 68198-5900
- Department of Pathology and Laboratory Medicine, King Fahad Specialist Hospital, Dammam, Saudi Arabia 31444
| | - Sugandha Saxena
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha NE 68198-5900
| | - Lingyun Wu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha NE 68198-5900
| | - Dipakkumar R. Prajapati
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha NE 68198-5900
| | - Abhilasha Purohit
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha NE 68198-5900
| | - Michelle L. Varney
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha NE 68198-5900
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE 68198-5870
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE 68198-5870
| | - Quan P. Ly
- Department of Surgical Oncology, University of Nebraska Medical Center, Omaha NE 68198-6880
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE 68198-5870
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE 68198-5870
| | - Rakesh K. Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha NE 68198-5900
| |
Collapse
|
40
|
Systemic but not MDSC-specific IRF4 deficiency promotes an immunosuppressed tumor microenvironment in a murine pancreatic cancer model. Cancer Immunol Immunother 2020; 69:2101-2112. [PMID: 32448983 PMCID: PMC7511276 DOI: 10.1007/s00262-020-02605-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 05/12/2020] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma is characterized by a strong immunosuppressive network with a dense infiltration of myeloid cells including myeloid-derived suppressor cells (MDSC). Two distinct populations of MDSC have been defined: polymorphonuclear MDSC (PMN-MDSC) and monocytic MDSC (M-MDSC). Several factors influence the development and function of MDSC including the transcription factor interferon regulatory factor 4 (IRF4). Here, we show that IRF4 deficiency accelerates tumor growth and reduces survival, accompanied with a dense tumor infiltration with PMN-MDSC and reduced numbers of CD8+ T cells. As IRF4 has been described to modulate myeloid cell development and function, particularly of PMN-MDSC, we analyzed its role using MDSC-specific IRF4 knockout mice with the Ly6G or LysM knock-in allele expressing Cre recombinase and Irf4flox. In GM-CSF-driven bone marrow cultures, IRF4 deficiency increased the frequency of MDSC-like cells with a strong T cell suppressive capacity. Myeloid (LysM)-specific depletion of IRF4 led to increased tumor weight and a moderate splenic M-MDSC expansion in tumor-bearing mice. PMN cell (Ly6G)-specific depletion of IRF4, however, did not influence tumor progression or MDSC accumulation in vivo in accordance with our finding that IRF4 is not expressed in PMN-MDSC. This study demonstrates a critical role of IRF4 in the generation of an immunosuppressive tumor microenvironment in pancreatic cancer, which is independent of IRF4 expression in PMN-MDSC.
Collapse
|
41
|
Liu Y, Feng M, Chen H, Yang G, Qiu J, Zhao F, Cao Z, Luo W, Xiao J, You L, Zheng L, Zhang T. Mechanistic target of rapamycin in the tumor microenvironment and its potential as a therapeutic target for pancreatic cancer. Cancer Lett 2020; 485:1-13. [PMID: 32428662 DOI: 10.1016/j.canlet.2020.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/21/2020] [Accepted: 05/04/2020] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer(PC) is a devastating disease with a poor prognosis; however, few treatment options are available and the search continues for feasible molecular therapeutic targets, both in the tumor itself and in the tumor microenvironment. The mechanistic target of rapamycin (mTOR) signaling pathway has emerged as an attractive target due to its regulatory role in multiple cellular processes, including metabolism, proliferation, survival, and differentiation, under physiological and pathological conditions. Although mTOR-regulated events in tumor cells and the tumor microenvironment are known to restrict the development and growth of tumor cells, monotherapy with mTOR inhibitors has shown limited efficacy against PC to date, suggesting the need for alternative approaches. In this review, we describe the mechanisms by which mTOR modulates the PC microenvironment and suggest ways its function in immune cells might be exploited for the treatment of PC. We also discuss preclinical and clinical studies with mTOR inhibitors in combination with other therapeutic strategies, most notably immunotherapy. Finally, we highlight the promise that mTOR combinatorial therapy may hold for the treatment of PC in the near future.
Collapse
Affiliation(s)
- Yueze Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Mengyu Feng
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China; Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Hao Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Fangyu Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Jianchun Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Clinical Immunology Center, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
42
|
Nalepa P, Gawecki R, Szewczyk G, Balin K, Dulski M, Sajewicz M, Mrozek-Wilczkiewicz A, Musioł R, Polanski J, Serda M. A [60]fullerene nanoconjugate with gemcitabine: synthesis, biophysical properties and biological evaluation for treating pancreatic cancer. Cancer Nanotechnol 2020. [DOI: 10.1186/s12645-020-00058-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Abstract
Background
The first-line chemotherapy drug that is used to treat pancreatic ductal adenocarcinoma is gemcitabine. Unfortunately, its effectiveness is hampered by its chemo-resistance, low vascularization and drug biodistribution limitations in the tumor microenvironment. Novel nanotherapeutics must be developed in order to improve the prognosis for patients with pancreatic cancer.
Results
We developed a synthetic methodology for obtaining a water-soluble nanoconjugate of a [60]fullerene-glycine derivative with the FDA-approved drug gemcitabine (nanoC60GEM). The proposed synthetic protocol enables a highly water-soluble [60]fullerene-glycine derivative (6) to be obtained, which was next successfully conjugated with gemcitabine using the EDCI/NHS carbodiimide protocol. The desired nanoconjugate was characterized using mass spectrometry and DLS, IR and XPS techniques. The photogeneration of singlet oxygen and the superoxide anion radical were studied by measuring 1O2 near-infrared luminescence at 1270 nm, followed by spin trapping of the DMPO adducts by EPR spectroscopy. The biological assays that were performed indicate that there is an inhibition of the cell cycle in the S phase and the induction of apoptosis by nanoC60GEM.
Conclusion
In this paper, we present a robust approach for synthesizing a highly water-soluble [60]fullerene nanoconjugate with gemcitabine. The performed biological assays on pancreatic cancer cell lines demonstrated cytotoxic effects of nanoC60GEM, which were enhanced by the generation of reactive oxygen species after blue LED irradiation of synthesized fullerene nanomaterial.
Collapse
|
43
|
Hu H, Tu W, Chen Y, Zhu M, Jin H, Huang T, Zou Z, Xia Q. The combination of PKM2 overexpression and M2 macrophages infiltration confers a poor prognosis for PDAC patients. J Cancer 2020; 11:2022-2031. [PMID: 32127930 PMCID: PMC7052945 DOI: 10.7150/jca.38981] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 12/26/2019] [Indexed: 01/22/2023] Open
Abstract
Macrophages play a critical role in the initiation and progression in various human solid tumors; however, their role and transformation in pancreatic ductal adenocarcinoma (PDAC) were still illusive. Here, immunohistochemistry was used to determine CD206 (specific marker of M2 macrophage) and PKM2 expression in PDAC tissues. Statistical analysis, such as Pearson χ2 test, Spearman's rank test, Kaplan-Meier and COX regression assay were used to evaluate their roles on PDAC prognosis. Data showed that both CD206 and PKM2 were elevated and responsible for a poor prognosis for PDAC. In addition, we showed that the two factors were positively correlated; co-overexpression of the two factors conferred the worst prognosis and functioned as an independent prognostic factor for the disease. Our data showed that M2 macrophage infiltration was correlated with PKM2 expression in PDAC cells. The two markers exerted synergistic effect on PDAC progression. Our results suggested dual-target inhibition M2 macrophage polarization and PKM2 expression of cancer cells might be novel approaches to treat PDAC.
Collapse
Affiliation(s)
- Hai Hu
- Department of Oncology, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai Cancer Institute, Shanghai, 200127, China
| | - Wenzhi Tu
- The Comprehensive Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Yungu Chen
- Department of Oncology, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai Cancer Institute, Shanghai, 200127, China
| | - Ming Zhu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 201620, China
| | - Huan Jin
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, Guangdong 510631, China
| | - Ting Huang
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, Guangdong 510631, China
| | - Zhengzhi Zou
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, Guangdong 510631, China
| | - Qing Xia
- Department of Oncology, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai Cancer Institute, Shanghai, 200127, China
| |
Collapse
|
44
|
The Microarchitecture of Pancreatic Cancer as Measured by Diffusion-Weighted Magnetic Resonance Imaging Is Altered by T Cells with a Tumor Promoting Th17 Phenotype. Int J Mol Sci 2020; 21:ijms21010346. [PMID: 31948053 PMCID: PMC6982276 DOI: 10.3390/ijms21010346] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 12/17/2022] Open
Abstract
Diffusion-weighted magnetic resonance imaging (DW-MRI) is a diagnostic tool that is increasingly used for the detection and characterization of focal masses in the abdomen, among these, pancreatic ductal adenocarcinoma (PDAC). DW-MRI reflects the microarchitecture of the tissue, and changes in diffusion, which are reflected by changes in the apparent diffusion coefficient (ADC), are mainly attributed to variations in cellular density, glandular formation, and fibrosis. When analyzing the T cell infiltrates, we found an association of a tumor-promoting subpopulation, characterized by the expression of interleukin (IL) 21 and IL26, with high ADC values. Moreover, the presence of IL21+ and IL26+ positive T cells was associated with poor prognosis. Pancreatic cancers—but not healthy pancreatic tissue—expressed receptors for IL21 and IL26, a finding that could be confirmed in pancreatic cell lines. The functionality of these receptors was demonstrated in pancreatic tumor cell lines, which showed phosphorylation of ERK1/2 and STAT3 pathways in response to the respective recombinant interleukins. Moreover, in vitro data showed an increased colony formation of tumor cells. In summary, our data showed an association of IL21+ and IL26+ immune cell infiltration, increased ADC, and aggressive tumor disease, most likely due to the activation of the key cancer signaling pathways ERK1/2 and STAT3 and formation of tumor colonies.
Collapse
|
45
|
Inflammation Associated Pancreatic Tumorigenesis: Upregulation of Succinate Dehydrogenase (Subunit B) Reduces Cell Growth of Pancreatic Ductal Epithelial Cells. Cancers (Basel) 2019; 12:cancers12010042. [PMID: 31877753 PMCID: PMC7016879 DOI: 10.3390/cancers12010042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 12/12/2019] [Accepted: 12/19/2019] [Indexed: 12/31/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is amongst the most fatal malignancies and its development is highly associated with inflammatory processes such as chronic pancreatitis (CP). Since the succinate dehydrogenase subunit B (SDHB) is regarded as tumor suppressor that is lost during cancer development, this study investigated the impact of M1-macrophages as part of the inflammatory microenvironment on the expression as well as function of SDHB in benign and premalignant pancreatic ductal epithelial cells (PDECs). Immunohistochemical analyses on pancreatic tissue sections from CP patients and control individuals revealed a stronger SDHB expression in ducts of CP tissues being associated with a greater abundance of macrophages compared to ducts in control tissues. Accordingly, indirect co-culture with M1-macrophages led to clearly elevated SDHB expression and SDH activity in benign H6c7-pBp and premalignant H6c7-kras PDECs. While siRNA-mediated SDHB knockdown in these cells did not affect glucose and lactate uptake after co-culture, SDHB knockdown significantly promoted PDEC growth which was associated with increased proliferation and decreased effector caspase activity particularly in co-cultured PDECs. Overall, these data indicate that SDHB expression and SDH activity are increased in PDECs when exposed to pro-inflammatory macrophages as a counterregulatory mechanism to prevent excessive PDEC growth triggered by the inflammatory environment.
Collapse
|
46
|
Interleukin 21 Receptor/Ligand Interaction Is Linked to Disease Progression in Pancreatic Cancer. Cells 2019; 8:cells8091104. [PMID: 31540511 PMCID: PMC6770770 DOI: 10.3390/cells8091104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/04/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) displays a marked fibro-inflammatory microenvironment in which infiltrated immune cells fail to eliminate the tumor cells and often—rather paradoxically—promote tumor progression. Of special interest are tumor-promoting T cells that assume a Th17-like phenotype because their presence in PDAC tissue is associated with a poor prognosis. In that context, the role of IL-21, a major cytokine released by Th17-like cells, was assessed. In all tissue samples (n = 264) IL-21+ immune cells were detected by immunohistochemistry and high density of those cells was associated with poor prognosis. In the majority of patients (221/264), tumor cells expressed the receptor for IL-21 (IL-21R) and also a downstream target of IL-21, Blimp-1 (199/264). Blimp-1 expression closely correlated with IL-21R expression and multivariate analysis revealed that expression of both IL-21R and Blimp-1 was associated with shorter survival time of the patients. In vitro data using pancreatic tumor cells lines provided a possible explanation: IL-21 activated ERK and STAT3 pathways and upregulated Blimp-1. Moreover, IL-21 increased invasion of tumor cell lines in a Blimp-1-dependent manner. As an in vivo correlate, an avian xenograft model was used. Here again Blimp-1 expression was significantly upregulated in IL-21 stimulated tumor cells. In summary, our data showed an association of IL-21+ immune cell infiltration and IL-21 receptor expression in PDAC with poor survival, most likely due to an IL-21-mediated promotion of tumor cell invasion and enhanced colony formation, supporting the notion of the tumor-promoting abilities of the tumor microenvironment.
Collapse
|
47
|
Yoshida S, Ito Z, Suka M, Bito T, Kan S, Akasu T, Saruta M, Okamoto M, Kitamura H, Fujioka S, Misawa T, Akiba T, Yanagisawa H, Sugiyama H, Koido S. Clinical Significance of Tumor-Infiltrating T Cells and Programed Death Ligand-1 in Patients with Pancreatic Cancer. Cancer Invest 2019; 37:463-477. [PMID: 31490702 DOI: 10.1080/07357907.2019.1661427] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The associations of the immunological status of the pancreatic ductal adenocarcinoma (PDA) microenvironment with prognosis were assessed. A high tumor-infiltrating lymphocyte (TIL) density was associated with a better prognosis. Importantly, even with a high density of TILs, the PDA cells with programed cell death-ligand 1 (PD-L1) expression showed a worse prognosis than the patients with negative PD-L1 expression. A significant association between a better prognosis and a tumor microenvironment with a high TIL density/negative PD-L1 expression was observed. Assessments of a combined immunological status in the tumor microenvironment may predict the prognosis of PDA patients following surgical resection.
Collapse
Affiliation(s)
- Sayumi Yoshida
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan
| | - Zensho Ito
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan
| | - Machi Suka
- Department of Public Health and Environmental Medicine, The Jikei University School of Medicine , Tokyo , Japan
| | - Tsuuse Bito
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan.,Institute of Clinical Medicine and Research, The Jikei University School of Medicine , Chiba , Japan
| | - Shin Kan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan.,Institute of Clinical Medicine and Research, The Jikei University School of Medicine , Chiba , Japan
| | - Takafumi Akasu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan
| | - Masayuki Saruta
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine , Tokyo , Japan
| | - Masato Okamoto
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine , Kanagawa , Japan
| | - Hiroaki Kitamura
- Department of Surgery, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan
| | - Shuichi Fujioka
- Department of Surgery, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan
| | - Takeyuki Misawa
- Department of Surgery, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan
| | - Tadashi Akiba
- Department of Surgery, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan
| | - Hiroyuki Yanagisawa
- Department of Public Health and Environmental Medicine, The Jikei University School of Medicine , Tokyo , Japan
| | - Haruo Sugiyama
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine , Osaka , Japan
| | - Shigeo Koido
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital , Chiba , Japan.,Institute of Clinical Medicine and Research, The Jikei University School of Medicine , Chiba , Japan
| |
Collapse
|
48
|
Fong CYK, Burke E, Cunningham D, Starling N. Up-to-Date Tailored Systemic Treatment in Pancreatic Ductal Adenocarcinoma. Gastroenterol Res Pract 2019; 2019:7135437. [PMID: 31582971 PMCID: PMC6748185 DOI: 10.1155/2019/7135437] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/09/2019] [Indexed: 12/24/2022] Open
Abstract
Despite intensive research efforts, pancreatic ductal adenocarcinoma is still regarded as an aggressive and life-limiting malignancy. Combination chemotherapy regimens that underpin the current treatment approach in the advanced setting have led to incremental survival gains in recent years but have failed to confer patients with a median overall survival that exceeds 12 months from diagnosis. Research has since focussed on understanding the role and interplay between various components of the desmoplastic stroma and tumour microenvironment, in addition to developing targeted therapies based on molecular features to improve the prognosis associated with this malignancy. This review will summarise the available systemic treatment options and discuss potential methods to refine the resolution of patient selection to enhance responses to currently available therapies. Furthermore, it will explore newer approaches anticipated to come to the fore of future clinical practice, such as agents targeting the DNA damage response and tumour microenvironment as well as immunotherapy-based combinations.
Collapse
Affiliation(s)
| | - Emma Burke
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM2 5PT, UK
| | - David Cunningham
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM2 5PT, UK
| | - Naureen Starling
- The Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM2 5PT, UK
| |
Collapse
|
49
|
Ray P, Ferraro M, Haag R, Quadir M. Dendritic Polyglycerol-Derived Nano-Architectures as Delivery Platforms of Gemcitabine for Pancreatic Cancer. Macromol Biosci 2019; 19:e1900073. [PMID: 31183964 DOI: 10.1002/mabi.201900073] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/07/2019] [Indexed: 12/14/2022]
Abstract
Dendritic polyglycerol-co-polycaprolactone (PG-co-PCL)-derived block copolymers are synthesized and explored as nanoscale drug delivery platforms for a chemotherapeutic agent, gemcitabine (GEM), which is the cornerstone of therapy for pancreatic ductal adenocarcinoma (PDAC). Current treatment strategies with GEM result in suboptimal therapeutic outcome owing to microenvironmental resistance and rapid metabolic degradation of GEM. To address these challenges, physicochemical and cell-biological properties of both covalently conjugated and non-covalently stabilized variants of GEM-containing PG-co-PCL architectures have been evaluated. Self-assembly behavior, drug loading and release capacity, cytotoxicity, and cellular uptake properties of these constructs in monolayer and in spheroid cultures of PDAC cells are investigated. To realize the covalently conjugated carrier systems, GEM, in conjunction with a tertiary amine, is attached to the polycarbonate block grafted from the PG-co-PCL core. It is observed that pH-dependent ionization properties of these amine side-chains direct the formation of self-assembly of block copolymers in the form of nanoparticles. For non-covalent encapsulation, a facile "solvent-shifting" technique is adopted. Fabrication techniques are found to control colloidal and cellular properties of GEM-loaded nanoconstructs. The feasibility and potential of these newly developed architectures for designing carrier systems for GEM to achieve augmented prognosis for pancreatic cancer are reported.
Collapse
Affiliation(s)
- Priyanka Ray
- Department of Coatings and Polymeric Materials, 1735 Research Park Drive, Fargo, ND, 58108-6050, USA
| | - Magda Ferraro
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195, Berlin, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195, Berlin, Germany
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric Materials, 1735 Research Park Drive, Fargo, ND, 58108-6050, USA
| |
Collapse
|
50
|
van Mackelenbergh MG, Stroes CI, Spijker R, van Eijck CHJ, Wilmink JW, Bijlsma MF, van Laarhoven HWM. Clinical Trials Targeting the Stroma in Pancreatic Cancer: A Systematic Review and Meta-Analysis. Cancers (Basel) 2019; 11:E588. [PMID: 31035512 PMCID: PMC6562438 DOI: 10.3390/cancers11050588] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 12/14/2022] Open
Abstract
The tumor microenvironment plays an important role in the initiation and progression of pancreatic adenocarcinoma (PDAC). In this systematic review, we provide an overview of clinical trials with stroma-targeting agents. We systematically searched MEDLINE/PubMed and the EMBASE database, using the PRISMA guidelines, for eligible clinical trials. In total, 2330 records were screened, from which we have included 106 articles. A meta-analysis could be performed on 51 articles which describe the targeting of the vascular endothelial growth factor (VEGF) pathway, and three articles which describe the targeting of hyaluronic acid. Anti-VEGF therapies did not show an increase in median overall survival (OS) with combined hazard ratios (HRs) of 1.01 (95% confidence interval (CI) 0.90-1.13). Treatment with hyaluronidase PEGPH20 showed promising results, but, thus far, only in combination with gemcitabine and nab-paclitaxel in selected patients with hyaluronic acid (HA)high tumors: An increase in median progression free survival (PFS) of 2.9 months, as well as a HR of 0.51 (95% CI 0.26-1.00). In conclusion, we found that anti-angiogenic therapies did not show an increased benefit in median OS or PFS in contrast to promising results with anti-hyaluronic acid treatment in combination with gemcitabine and nab-paclitaxel. The PEGPH20 clinical trials used patient selection to determine eligibility based on tumor biology, which underlines the importance to personalize treatment for pancreatic cancer patients.
Collapse
Affiliation(s)
- Madelaine G van Mackelenbergh
- Laboratory of Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Charlotte I Stroes
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - René Spijker
- Medical Library, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
- Cochrane Netherlands, Julius Center, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands.
| | - Casper H J van Eijck
- Department of Surgery, Erasmus MC, Dr. Molewaterplein 40, 3015GD Rotterdam, The Netherlands.
| | - Johanna W Wilmink
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Maarten F Bijlsma
- Laboratory of Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| |
Collapse
|