1
|
Pele KG, Calderón-Villalba A, Amaveda H, Mora M, Zhang-Zhou J, Pérez MÁ, García-Aznar JM, Alamán-Díez P, García-Gareta E. Novel hydrogel-based cancer-on-a-chip models for growth of 3D multi-cellular structures and investigation of early angiogenesis in pancreatic ductal adenocarcinoma. Colloids Surf B Biointerfaces 2025; 253:114736. [PMID: 40315572 DOI: 10.1016/j.colsurfb.2025.114736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/27/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
Cancer-on-a-chip models have enormous potential for the study of tumour development events. Here, we investigated hydrogels of egg white (EW) and gelatin for growth of 3D multi-cellular structures and investigation of early angiogenesis inside microfluidic devices. We focused on pancreatic ductal adenocarcinoma (PDAC), a devastating gastrointestinal malignancy. EW/gelatin hydrogels were stiffer and showed porous globular structures compared to the fibrous network of collagen I molecules. PANC-1 cells preferentially formed significantly larger spheroids in collagen I than in EW/gelatin hydrogels, whilst cell aggregates in the shape of grape-like clusters were significantly larger and more abundant in EW/gelatin. Cells inside the aggregates showed active cell unions, secreted matrix, and formed active unions with the surrounding EW/gelatin hydrogel. Early stages of PDAC were recreated by co-culture of two different microenvironments, one for PANC-1 and another one for fibroblasts, for investigating the secretion of soluble angiogenic factors, which depended on the role of each factor in the angiogenic and tumorigenic processes. Overall, cancer cell proliferation and establishment of a tumour vasculature were favoured. This study demonstrates the importance of the microenvironment in tumour cells behaviour as well as the complex interplay between the different cells present in PDAC to establish a tumoural vasculature.
Collapse
Affiliation(s)
- Karinna Georgiana Pele
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Alejandro Calderón-Villalba
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Hippolyte Amaveda
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC and University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Mario Mora
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC and University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Jack Zhang-Zhou
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - María Ángeles Pérez
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain
| | - José Manuel García-Aznar
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain
| | - Pilar Alamán-Díez
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Elena García-Gareta
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain; Division of Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, University College London, London NW3 2QG, United Kingdom.
| |
Collapse
|
2
|
Ulus G, Özbek EN, Yılmaz H, Keselik E, Sarıcaoğlu M, Akyol Bahçeci S, İşel E, Debeleç Bütüner B, Yetik Anacak G, Koparal AT. Borax pentahydrate as a promising boron-based angiogenesis inhibitor. J Trace Elem Med Biol 2025; 89:127640. [PMID: 40203787 DOI: 10.1016/j.jtemb.2025.127640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Boron, a trace element, is involved in various physiological and metabolic processes, and recent studies suggest that boron compounds may have potential in cancer prevention and treatment. In this study, the antiangiogenic effects of a boron compound, borax pentahydrate (BPH), were investigated. Angiogenesis is a tightly regulated biological process responsible for the formation of new blood vessels from existing vasculatures. This process plays a critical role in cancer progression, making it an important target for cancer therapy. Pancreatic and kidney cancers are difficult to treat because they are aggressive and resistant to chemotherapy. METHODS The antiproliferative activity was evaluated using the MTT assay, while antiangiogenic effects were tested through in vitro tube formation assays and in ovo chick chorioallantoic membrane (CAM) assay. The effect of BPH on VEGF levels was determined using Western blot analysis in HUVEC, ACHN, PANC-1 cells. The effect of BPH on tumor angiogenesis was investigated with an in vivo Ehrlich ascites carcinoma model (EAC). RESULTS BPH exhibited potent antiproliferative and antiangiogenic activities, inhibiting the proliferation of ACHN, PANC-1, and HUVECs, disrupting endothelial tube formation, and inhibiting vascular formation on the CAM surface in a dose-dependent manner. VEGF levels were significantly decreased in ACHN, PANC-1 and HUVECs. There was also a decrease in VEGF and TGF-β1 levels in BPH-treated tumor groups. In addition, BPH caused a decrease in tumor size. CONCLUSION These findings suggest that BPH may be a new antiangiogenic and antitumoral agent. BPH may contribute to drug development studies targeting angiogenesis-related diseases as a promising new therapeutic agent.
Collapse
Affiliation(s)
- G Ulus
- Republic of Türkiye, Ministry of Education, Şerife Bacı Vocational and Technical High School, Izmir 35090, Turkiye.
| | - E N Özbek
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkiye
| | - H Yılmaz
- Republic of Türkiye, Ministry of Education, Mimar Sinan Vocational and Technical High School, Izmir 35090, Turkiye
| | - E Keselik
- Department of Histology and Embryology, Faculty of Medicine, Katip Çelebi University, Izmir 35100, Turkiye
| | - M Sarıcaoğlu
- Department of Histology and Embryology, Faculty of Medicine, Katip Çelebi University, Izmir 35100, Turkiye
| | - S Akyol Bahçeci
- Department of Histology and Embryology, Faculty of Medicine, Katip Çelebi University, Izmir 35100, Turkiye
| | - E İşel
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkiye
| | - B Debeleç Bütüner
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkiye
| | - G Yetik Anacak
- Department of Pharmacology, Faculty of Pharmacy, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkiye
| | - A T Koparal
- Yunus Emre Vocational School of Health Services, Anadolu University, Eskisehir 26470, Turkiye
| |
Collapse
|
3
|
Liu Q, Li R, Zhu W, Zheng P. Case report: Microsatellite instability-high pancreas adenosquamous carcinoma with postoperative liver metastasis recurrence treated with multimodality therapy achieving complete pathological response. Front Immunol 2024; 15:1456343. [PMID: 39726603 PMCID: PMC11669589 DOI: 10.3389/fimmu.2024.1456343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Pancreatic adenosquamous carcinoma (PASC) is a rare subtype of pancreatic cancer (PC), with no established consensus on the optimal treatment for postoperative liver metastasis recurrence. We report a case of a 48-year-old male patient who underwent radical surgery and was pathologically diagnosed with microsatellite instability-high (MSI-H) PASC. The patient experienced liver metastasis recurrence following single-agent gemcitabine adjuvant chemotherapy. After one session of transarterial chemoembolization (TACE) with oxaliplatin, fluorouracil, and epirubicin, followed by six cycles of adjuvant chemotherapy with gemcitabine and nab-paclitaxel combined with sintilimab immunotherapy and bevacizumab targeted therapy, complete pathological regression of the liver metastasis was achieved. The patient has now reached a 24-month survival period and continues to be monitored at our center. This case illustrates the promise of the proposed treatment regimen, highlighting the significant potential of multimodality strategies in managing metastatic recurrence of MSI-H PASC.
Collapse
Affiliation(s)
- Qinghua Liu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Ruoyun Li
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Wei Zhu
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, China
| | - Pengfei Zheng
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
4
|
Yousefi T, Mohammadi Jobani B, Taebi R, Qujeq D. Innovating Cancer Treatment Through Cell Cycle, Telomerase, Angiogenesis, and Metastasis. DNA Cell Biol 2024; 43:438-451. [PMID: 39018567 DOI: 10.1089/dna.2024.0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/19/2024] Open
Abstract
Cancer remains a formidable challenge in the field of medicine, necessitating innovative therapeutic strategies to combat its relentless progression. The cell cycle, a tightly regulated process governing cell growth and division, plays a pivotal role in cancer development. Dysregulation of the cell cycle allows cancer cells to proliferate uncontrollably. Therapeutic interventions designed to disrupt the cell cycle offer promise in restraining tumor growth and progression. Telomerase, an enzyme responsible for maintaining telomere length, is often overactive in cancer cells, conferring them with immortality. Targeting telomerase presents an opportunity to limit the replicative potential of cancer cells and hinder tumor growth. Angiogenesis, the formation of new blood vessels, is essential for tumor growth and metastasis. Strategies aimed at inhibiting angiogenesis seek to deprive tumors of their vital blood supply, thereby impeding their progression. Metastasis, the spread of cancer cells from the primary tumor to distant sites, is a major challenge in cancer therapy. Research efforts are focused on understanding the underlying mechanisms of metastasis and developing interventions to disrupt this deadly process. This review provides a glimpse into the multifaceted approach to cancer therapy, addressing critical aspects of cancer biology-cell cycle regulation, telomerase activity, angiogenesis, and metastasis. Through ongoing research and innovative strategies, the field of oncology continues to advance, offering new hope for improved treatment outcomes and enhanced quality of life for cancer patients.
Collapse
Affiliation(s)
- Tooba Yousefi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahareh Mohammadi Jobani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reyhaneh Taebi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Durdi Qujeq
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
5
|
Sweeney A, Xavierselvan M, Langley A, Solomon P, Arora A, Mallidi S. Vascular regional analysis unveils differential responses to anti-angiogenic therapy in pancreatic xenografts through macroscopic photoacoustic imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.27.595784. [PMID: 38854042 PMCID: PMC11160648 DOI: 10.1101/2024.05.27.595784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy and the third leading cause of cancer deaths in the U.S. Despite major innovations in imaging technologies, there are limited surrogate radiographic indicators to aid in therapy planning and monitoring. Amongst the various imaging techniques Ultrasound-guided photoacoustic imaging (US-PAI) is a promising modality based on endogenous blood (hemoglobin) and blood oxygen saturation (StO 2 ) contrast to monitor response to anti-angiogenic therapies. Adaptation of US-PAI to the clinical realm requires macroscopic configurations for adequate depth visualization, illuminating the need for surrogate radiographic markers, including the tumoral microvessel density (MVD). In this work, subcutaneous xenografts with PC cell lines AsPC-1 and MIA-PaCa-2 were used to investigate the effects of receptor tyrosine kinase inhibitor (sunitinib) treatment on MVD and StO 2 . Through histological correlation, we have shown that regions of high and low vascular density (HVD and LVD) can be identified through frequency domain filtering of macroscopic PA images which could not be garnered from purely global analysis. We utilized vascular regional analysis (VRA) of treatment-induced StO 2 and total hemoglobin (HbT) changes. VRA as a tool to monitor treatment response allowed us to identify potential timepoints of vascular remodeling, highlighting its ability to provide insights into the TME not only for sunitinib treatment but also other anti-angiogenic therapies.
Collapse
|
6
|
Wen J. Single-cell transcriptomics reveals antigen-presenting capacity and therapeutic resistance potential of immunomodulatory endothelial cells in colorectal cancer. Immun Inflamm Dis 2024; 12:e1311. [PMID: 38874280 PMCID: PMC11177288 DOI: 10.1002/iid3.1311] [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: 11/24/2023] [Revised: 05/10/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND The heterogeneity of tumor endothelial cells (TECs) hinders the efficacy of antiangiogenic therapies (AATs). Only a small percentage of angiogenic TECs are considered effective targets for AATs. Immunomodulatory ECs (IMECs), as a newly focused functional subgroup of endothelial cells (ECs), are being evaluated for their ability to regulate tumor immune balance and influence existing AATs. METHODS Based on single-cell transcriptome data from colorectal cancer in a publicly available database, we conducted a wide array of bioinformatic approaches to study EC subsets that meet the IMECs definition. Our investigation encompassed the gene expression signatures of these subsets, cellular composition differences, cell-cell interactions. RESULTS Two subsets that meet the IMECs definition were found in tumors and para-cancerous tissues. Combined with the results of gene ontological analysis and interaction with CD4+ T cells, we found that IMECs can present MHC-II antigens to mature CD4+ T cells. There were differences in the level of interaction between IMECs and different types of mature CD4+ T cell subsets. In addition, IMEC subsets had different expression levels of angiogenesis related genes. The angiogenesis score of IMECs decreased after patients received immunotherapy. IMEC subsets do not depend on a single proangiogenic receptor and are involved in regulating angiogenesis, which may reduce the efficacy of AATs. The adverse effects of specific IMEC subsets on AATs were validated in the RNA-seq dataset of the bevacizumab treatment group. CONCLUSION Our study suggests the potential MHC-II antigen presentation capacity of IMECs and the enhanced angiogenesis characteristics within tumors. The function of IMECs in the vascular network may have a potentially adverse effect on AATs. Controlling the functional properties of IMECs may be a new angle for tumor therapy.
Collapse
Affiliation(s)
- Jingyi Wen
- School of Biology and Biological EngineeringSouth China University of TechnologyGuangzhouGuangdongChina
| |
Collapse
|
7
|
Shinde A, Panchal K, Patra P, Singh S, Enakolla S, Paliwal R, Chaurasiya A. QbD Enabled Development and Evaluation of Pazopanib Loaded Nanoliposomes for PDAC Treatment. AAPS PharmSciTech 2024; 25:97. [PMID: 38710894 DOI: 10.1208/s12249-024-02806-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the highly fatal types of cancer with high mortality/incidence. Considering the crucial role of vascular endothelial growth factor (VEGF) in PDAC progression, its inhibition can be a viable strategy for the treatment. Pazopanib, a second-generation VEGF inhibitor, is approved for the treatment of various oncological conditions. However, due to associated limitations like low oral bioavailability (14-39%), high inter/intra-subject variability, stability issues, etc., high doses (800 mg) are required, which further lead to non-specific toxicities and also contribute toward cancer resistance. Thus, to overcome these challenges, pazopanib-loaded PEGylated nanoliposomes were developed and evaluated against pancreatic cancer cell lines. The nanoliposomes were prepared by thin-film hydration method, followed by characterization and stability studies. This QbD-enabled process design successfully led to the development of a suitable pazopanib liposomal formulation with desirable properties. The % entrapment of PZP-loaded non-PEGylated and PEGylated nanoliposomes was found to be 75.2% and 84.9%, respectively, whereas their particle size was found to be 129.7 nm and 182.0 nm, respectively. The developed liposomal formulations exhibited a prolonged release and showed desirable physicochemical properties. Furthermore, these liposomal formulations were also assessed for in vitro cell lines, such as cell cytotoxicity assay and cell uptake. These studies confirm the effectiveness of developed liposomal formulations against pancreatic cancer cell lines. The outcomes of this work provide encouraging results and a way forward to thoroughly investigate its potential for PDAC treatment.
Collapse
Affiliation(s)
- Aishwarya Shinde
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Kanan Panchal
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Parameswar Patra
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Sonali Singh
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Sucharitha Enakolla
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Rishi Paliwal
- Nanomedicine and Bioengineering Research Laboratory, Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | - Akash Chaurasiya
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India.
| |
Collapse
|
8
|
Ezelarab HAA, Abd El-Hafeez AA, Ali TFS, Sayed AM, Hassan HA, Beshr EAM, Abbas SH. New 2-oxoindole derivatives as multiple PDGFRα/ß and VEGFR-2 tyrosine kinase inhibitors. Bioorg Chem 2024; 145:107234. [PMID: 38412650 DOI: 10.1016/j.bioorg.2024.107234] [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: 10/24/2023] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 02/29/2024]
Abstract
Two new series of N-aryl acetamides 6a-o and benzyloxy benzylidenes 9a-p based 2-oxoindole derivatives were designed as potent antiproliferative multiple kinase inhibitors. The results of one-dose NCI antiproliferative screening for compounds 6a-o and 9a-p elucidated that the most promising antiproliferative scaffolds were 6f and 9f, which underwent five-dose testing. Notably, the amido congener 6f was the most potent derivative towards pancreatic ductal adenocarcinoma MDA-PATC53 and PL45 cell lines (IC50 = 1.73 µM and 2.40 µM, respectively), and the benzyloxy derivative 9f was the next potent one with IC50 values of 2.85 µM and 2.96 µM, respectively. Both compounds 6f and 9f demonstrated a favorable safety profile when tested against normal prostate epithelial cells (RWPE-1). Additionally, compound 6f displayed exceptional selectivity as a multiple kinase inhibitor, particularly targeting PDGFRα, PDGFRβ, and VEGFR-2 kinases, with IC50 values of 7.41 nM, 6.18 nM, and 7.49 nM, respectively. In contrast, the reference compound Sunitinib exhibited IC50 values of 43.88 nM, 2.13 nM, and 78.46 nM against the same kinases. The derivative 9f followed closely, with IC50 values of 9.9 nM, 6.62 nM, and 22.21 nM for the respective kinases. Both 6f and 9f disrupt the G2/M cell cycle transition by upregulating p21 and reducing CDK1 and cyclin B1 mRNA levels. The interplay between targeted kinases and these cell cycle regulators underpins the G2/M cell cycle arrest induced by our compounds. Also, compounds 6f and 9f fundamentally resulted in entering MDA-PATC53 cells into the early stage of apoptosis with good percentages compared to the positive control Sunitinib. The in silico molecular-docking outcomes of scaffolds 6a-o and 9a-p in VEGFR-2, PDGFRα, and PDGFRβ active sites depicted their ability to adopt essential binding interactions like the reference Sunitinib. Our designed analogs, specifically 6f and 9f, possess promising antiproliferative and kinase inhibitory properties, making them potential candidates for further therapeutic development.
Collapse
Affiliation(s)
- Hend A A Ezelarab
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Amer Ali Abd El-Hafeez
- Pharmacology and Experimental Oncology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Taha F S Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Ahmed M Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University, 62513 Beni-Suef, Egypt; Department of Pharmacognosy, Collage of Pharmacy, Almaaqal University, 61014 Basrah, Iraq
| | - Heba A Hassan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt.
| | - Eman A M Beshr
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Samar H Abbas
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt.
| |
Collapse
|
9
|
Khafaga AF, Gaballa MMS, Karam R, Shoulah SA, Shamma RN, Khalifa NE, Farrag NE, Noreldin AE. Synergistic therapeutic strategies and engineered nanoparticles for anti-vascular endothelial growth factor therapy in cancer. Life Sci 2024; 341:122499. [PMID: 38342375 DOI: 10.1016/j.lfs.2024.122499] [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: 10/15/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/13/2024]
Abstract
Angiogenesis is one of the defining characteristics of cancer. Vascular endothelial growth factor (VEGF) is crucial for the development of angiogenesis. A growing interest in cancer therapy is being caused by the widespread use of antiangiogenic drugs in treating several types of human cancer. However, this therapeutic approach can worsen resistance, invasion, and overall survival. As we proceed, refining combination strategies and addressing the constraint of targeted treatments are paramount. Therefore, major challenges in using novel combinations of antiangiogenic agents with cytotoxic treatments are currently focused on illustrating the potential of synergistic therapeutic strategies, alongside advancements in nanomedicine and gene therapy, present opportunities for more precise interference with angiogenesis pathways and tumor environments. Nanoparticles have the potential to regulate several crucial activities and improve several drug limitations such as lack of selectivity, non-targeted cytotoxicity, insufficient drug delivery at tumor sites, and multi-drug resistance based on their unique features. The goal of this updated review is to illustrate the enormous potential of novel synergistic therapeutic strategies and the targeted nanoparticles as an alternate strategy for t treating a variety of tumors employing antiangiogenic therapy.
Collapse
Affiliation(s)
- Asmaa F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina 22758, Egypt.
| | - Mohamed M S Gaballa
- Department of Pathology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt.
| | - Reham Karam
- Department of Virology, Faculty of Veterinary Medicine, Mansoura University, 35511, Egypt.
| | - Salma A Shoulah
- Department of Animal Medicine (Infectious Diseases), Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt.
| | - Rehab N Shamma
- Department of Pharmaceutics & Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Norhan E Khalifa
- Department of Physiology, Faculty of Veterinary Medicine, Matrouh University, Matrouh 51511, Egypt.
| | - Nehal E Farrag
- Faculty of Veterinary Medicine, Alexandria University, Edfina 22758, Egypt.
| | - Ahmed E Noreldin
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt.
| |
Collapse
|
10
|
Wu J, Liu Y, Fu Q, Cao Z, Ma X, Li X. Characterization of tumor-associated endothelial cells and the development of a prognostic model in pancreatic ductal adenocarcinoma. Biochim Biophys Acta Gen Subj 2024; 1868:130545. [PMID: 38141886 DOI: 10.1016/j.bbagen.2023.130545] [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: 10/18/2023] [Revised: 12/10/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy characterized by a complex tumor microenvironment. Angiogenesis is of paramount importance in the proliferation and metastasis of PDAC. However, currently, there are no well-defined biomarkers available to guide the prognosis and treatment of PDAC. In this study, we investigated the interactions between tumor-associated endothelial cells (TAECs) and tumor cells in PDAC, and identified a specific subset of TAECs characterized by high expression of COL4A1. COL4A1+ endothelial cells interact with tumor cells through the COLLAGEN signaling pathway to promote tumor cell proliferation, migration, and invasion. We also observed activation of HOXD9 in COL4A1+ endothelial cells. Based on these findings, we developed a prognostic model called TaEMS, which accurately predicts patient prognosis. TaEMS identified high-risk patients enriched in cell cycle-related pathways and low-risk patients enriched in focal adhesions, smooth muscle regulation, and immune pathways. Moreover, high-risk patients displayed a reduced level of immune cell infiltration, indicating the presence of a "cold tumor" phenotype. Overall, our study uncovered an intricate crosstalk between TAECs and tumor cells in PDAC, emphasizing the role of HOXD9 and highlighting the potential of TaEMS as a prognostic biomarker for precise therapies.
Collapse
Affiliation(s)
- Jun Wu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443003, China; Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong; Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Yang Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qi Fu
- Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong; Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Zhi Cao
- Department of Gastroenterology, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518020, Guangdong Province, China
| | - Xiaodong Ma
- Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong; Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China.
| | - Xun Li
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443003, China.
| |
Collapse
|
11
|
Denda Y, Matsuo Y, Sugita S, Eguchi Y, Nonoyama K, Murase H, Kato T, Imafuji H, Saito K, Morimoto M, Ogawa R, Takahashi H, Mitsui A, Kimura M, Takiguchi S. The Natural Product Parthenolide Inhibits Both Angiogenesis and Invasiveness and Improves Gemcitabine Resistance by Suppressing Nuclear Factor κB Activation in Pancreatic Cancer Cell Lines. Nutrients 2024; 16:705. [PMID: 38474833 PMCID: PMC10934733 DOI: 10.3390/nu16050705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/18/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
We previously established pancreatic cancer (PaCa) cell lines resistant to gemcitabine and found that the activity of nuclear factor κB (NF-κB) was enhanced upon the acquisition of gemcitabine resistance. Parthenolide, the main active ingredient in feverfew, has been reported to exhibit antitumor activity by suppressing the NF-κB signaling pathway in several types of cancers. However, the antitumor effect of parthenolide on gemcitabine-resistant PaCa has not been elucidated. Here, we confirmed that parthenolide significantly inhibits the proliferation of both gemcitabine-resistant and normal PaCa cells at concentrations of 10 µM and higher, and that the NF-κB activity is significantly inhibited, even by 1 µM parthenolide. In Matrigel invasion assays and angiogenesis assays, the invasive and angiogenic potentials were higher in gemcitabine-resistant than normal PaCa cells and were inhibited by a low concentration of parthenolide. Furthermore, Western blotting showed suppressed MRP1 expression in gemcitabine-resistant PaCa treated with a low parthenolide concentration. In a colony formation assay, the addition of 1 µM parthenolide improved the sensitivity of gemcitabine-resistant PaCa cell lines to gemcitabine. These results suggest that parthenolide may be used as a novel therapeutic agent for the treatment of gemcitabine-resistant PaCa.
Collapse
Affiliation(s)
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (Y.D.); (S.S.); (Y.E.); (K.N.); (H.M.); (T.K.); (H.I.); (K.S.); (M.M.); (R.O.); (H.T.); (A.M.); (M.K.); (S.T.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Fard D, Giraudo E, Tamagnone L. Mind the (guidance) signals! Translational relevance of semaphorins, plexins, and neuropilins in pancreatic cancer. Trends Mol Med 2023; 29:817-829. [PMID: 37598000 DOI: 10.1016/j.molmed.2023.07.009] [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: 06/24/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 08/21/2023]
Abstract
Pancreatic cancer is a major cause of demise worldwide. Although key associated genetic changes have been discovered, disease progression is sustained by pathogenic mechanisms that are poorly understood at the molecular level. In particular, the tissue microenvironment of pancreatic adenocarcinoma (PDAC) is usually characterized by high stromal content, scarce recruitment of immune cells, and the presence of neuronal fibers. Semaphorins and their receptors, plexins and neuropilins, comprise a wide family of regulatory signals that control neurons, endothelial and immune cells, embryo development, and normal tissue homeostasis, as well as the microenvironment of human tumors. We focus on the role of these molecular signals in pancreatic cancer progression, as revealed by experimental research and clinical studies, including novel approaches for cancer treatment.
Collapse
Affiliation(s)
- Damon Fard
- Università Cattolica del Sacro Cuore, Department of Life Sciences and Public Health, Rome, Italy
| | - Enrico Giraudo
- Department of Science and Drug Technology, University of Turin, Turin, Italy; Candiolo Cancer Institute, FPO IRCCS, Candiolo, Turin, Italy
| | - Luca Tamagnone
- Università Cattolica del Sacro Cuore, Department of Life Sciences and Public Health, Rome, Italy; Fondazione Policlinico Gemelli, IRCCS, Rome, Italy.
| |
Collapse
|
13
|
Li Y, Yang KD, Duan HY, Du YN, Ye JF. Phage-based peptides for pancreatic cancer diagnosis and treatment: alternative approach. Front Microbiol 2023; 14:1231503. [PMID: 37601380 PMCID: PMC10433397 DOI: 10.3389/fmicb.2023.1231503] [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: 05/30/2023] [Accepted: 07/06/2023] [Indexed: 08/22/2023] Open
Abstract
Pancreatic cancer is a devastating disease with a high mortality rate and a lack of effective therapies. The challenges associated with early detection and the highly aggressive nature of pancreatic cancer have limited treatment options, underscoring the urgent need for better disease-modifying therapies. Peptide-based biotherapeutics have become an attractive area of research due to their favorable properties such as high selectivity and affinity, chemical modifiability, good tissue permeability, and easy metabolism and excretion. Phage display, a powerful technique for identifying peptides with high affinity and specificity for their target molecules, has emerged as a key tool in the discovery of peptide-based drugs. Phage display technology involves the use of bacteriophages to express peptide libraries, which are then screened against a target of interest to identify peptides with desired properties. This approach has shown great promise in cancer diagnosis and treatment, with potential applications in targeting cancer cells and developing new therapies. In this comprehensive review, we provide an overview of the basic biology of phage vectors, the principles of phage library construction, and various methods for binding affinity assessment. We then describe the applications of phage display in pancreatic cancer therapy, targeted drug delivery, and early detection. Despite its promising potential, there are still challenges to be addressed, such as optimizing the selection process and improving the pharmacokinetic properties of phage-based drugs. Nevertheless, phage display represents a promising approach for the development of novel targeted therapies in pancreatic cancer and other tumors.
Collapse
Affiliation(s)
- Yang Li
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Kai-di Yang
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Hao-yu Duan
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Ya-nan Du
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Jun-feng Ye
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| |
Collapse
|
14
|
Hadi MM, Farrell S, Nesbitt H, Thomas K, Kubajewska I, Ng A, Masood H, Patel S, Sciscione F, Davidson B, Callan JF, MacRobert AJ, McHale AP, Nomikou N. Nanotechnology-augmented sonodynamic therapy and associated immune-mediated effects for the treatment of pancreatic ductal adenocarcinoma. J Cancer Res Clin Oncol 2023; 149:5007-5023. [PMID: 36319895 PMCID: PMC10349707 DOI: 10.1007/s00432-022-04418-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/12/2022] [Indexed: 03/23/2023]
Abstract
PURPOSE Sonodynamic therapy (SDT) is emerging as a cancer treatment alternative with significant advantages over conventional therapies, including its minimally invasive and site-specific nature, its radical antitumour efficacy with minimal side effects, and its capacity to raise an antitumour immune response. The study explores the efficacy of SDT in combination with nanotechnology against pancreatic ductal adenocarcinoma. METHODS A nanoparticulate formulation (HPNP) based on a cathepsin B-degradable glutamate-tyrosine co-polymer that carries hematoporphyrin was used in this study for the SDT-based treatment of PDAC. Cathepsin B levels in BxPC-3 and PANC-1 cells were correlated to cellular uptake of HPNP. The HPNP efficiency to induce a sonodynamic effect at varying ultrasound parameters, and at different oxygenation and pH conditions, was investigated. The biodistribution, tumour accumulation profile, and antitumour efficacy of HPNP in SDT were examined in immunocompetent mice carrying bilateral ectopic murine pancreatic tumours. The immune response profile of excised tumour tissues was also examined. RESULTS The HPNP formulation significantly improved cellular uptake of hematoporphyrin for both BxPC-3 and PANC-1 cells, while increase of cellular uptake was positively correlated in PANC-1 cells. There was a clear SDT-induced cytotoxicity at the ultrasound conditions tested, and the treatment impaired the capacity of both BxPC-3 and PANC-1 cells to form colonies. The overall acoustic energy and pulse length, rather than the power density, were key in eliciting the effects observed in vitro. The SDT treatment in combination with HPNP resulted in 21% and 27% reduction of the target and off-target tumour volumes, respectively, within 24 h. A single SDT treatment elicited an antitumour effect that was characterized by an SDT-induced decrease in immunosuppressive T cell phenotypes. CONCLUSION SDT has significant potential to serve as a monotherapy or adjunctive treatment for inoperable or borderline resectable PDAC.
Collapse
Affiliation(s)
- Marym Mohammad Hadi
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | - Sian Farrell
- Biomedical Sciences Research Institute, Ulster University, Coleraine, UK
| | - Heather Nesbitt
- Biomedical Sciences Research Institute, Ulster University, Coleraine, UK
| | - Keith Thomas
- Biomedical Sciences Research Institute, Ulster University, Coleraine, UK
| | - Ilona Kubajewska
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
- Nanomerics Ltd, London, UK
| | - Alex Ng
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | - Hamzah Masood
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | - Shiv Patel
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | - Fabiola Sciscione
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | - Brian Davidson
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | - John F Callan
- Biomedical Sciences Research Institute, Ulster University, Coleraine, UK
| | - Alexander J MacRobert
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | - Anthony P McHale
- Biomedical Sciences Research Institute, Ulster University, Coleraine, UK
| | - Nikolitsa Nomikou
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK.
| |
Collapse
|
15
|
Zhang D, Tang W, Shen X, Weng S, Dong L. Comprehensive Investigation Illustrates the Role of M2 Macrophages and Its Related Genes in Pancreatic Cancer. Medicina (B Aires) 2023; 59:medicina59040717. [PMID: 37109675 PMCID: PMC10146353 DOI: 10.3390/medicina59040717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023] Open
Abstract
Background and Objectives: M2 macrophages play an important role in cancers. Our study aimed to illustrate the effect of M2 macrophages in pancreatic cancer (PC). Materials and Methods: The open-access data used for analysis were downloaded from The Cancer Genome Atlas Program database, as well as some online databases. R software was mainly used for data analysis based on the specific packages. Results: Here, we comprehensively investigated the role of M2 macrophages and their related genes in PC. We performed the biological enrichment of M2 macrophages in PC. Meanwhile, we identified adenosine A3 receptor (TMIGD3) as the interest gene for further analysis. The single-cell analysis showed that was mainly expressed in the Mono/Macro cells based on the data from multiple data cohorts. Biological investigation showed that TMIGD3 was primarily enriched in angiogenesis, pancreas-beta cells and TGF-beta signaling. Tumor microenvironment analysis indicated that TMIGD3 was positively correlated with monocyte_MCPCOUNTER, NK cell_MCPCOUNTER, macrophages M2_CIBERSORT, macrophage_EPIC, neutrophil_TIMER and endothelial cell_MCPCOUNTER. Interestingly, we determined that all the immune functions quantified by single sample gene set enrichment analysis algorithms were activated in the patients with high TMIGD3 expression. Conclusions: Our results provide a novel direction for the research focused on the M2 macrophages in PC. Meanwhile, TMIGD3 was identified as an M2 macrophage-related biomarker for PC.
Collapse
Affiliation(s)
- Danying Zhang
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wenqing Tang
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xizhong Shen
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shuqiang Weng
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
16
|
Involvement of angiogenesis in cancer-associated acinar-to-ductal metaplasia lesion of pancreatic cancer invasive front. J Cancer Res Clin Oncol 2023:10.1007/s00432-022-04554-5. [PMID: 36592214 DOI: 10.1007/s00432-022-04554-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 12/21/2022] [Indexed: 01/03/2023]
Abstract
PURPOSE This study aimed to demonstrate the involvement of angiogenesis in cancer-associated acinar-to-ductal metaplasia (CA-ADM) lesion of invasive front pancreatic ductal adenocarcinoma (PDAC) and investigate the possible mechanism. METHODS Tissue samples from 128 patients with PDAC and 36 LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx-1-Cre mice were analyzed. Immunohistochemical assay was performed using HE, anti-CK19 and anti-amylase to confirm the presence of CA-ADM lesions, using anti-CD34 and anti-CD31 to measure microvessel density (MVD), and using anti-CD68, anti-CD163, anti-iNOS, or anti-MMP9 to evaluate the immune microenvironment. We performed multiplex immunohistochemical assay to detect the co-expression of MMP9 and CD68 on macrophage. We examined clinical outcomes and other clinicopathological factors to determine the significance of high-level MVD of CA-ADM on survival and liver metastasis. We performed tube formation assay to evaluate the effect of macrophage on angiogenic capacity in vitro. RESULTS Angiogenesis was significantly abundant in CA-ADM lesions compared with that in PDAC lesions in human and mouse tissues. High-level MVD in CA-ADM lesions was an independent predictor of poor prognosis (P = 0.0047) and the recurrence of liver metastasis (P = 0.0027). More CD68-positive and CD163-positive macrophages were detected in CA-ADM lesions than in PDAC. The percentage of CD68-positive macrophages was positively correlated with MVD in CA-ADM lesions. Multiplex-immunostaining revealed that MMP9 was expressed in CD68-positive macrophages of CA-ADM lesions. In CA-ADM lesions, the percentage of macrophages was positively correlated with MMP9 expression, which positively correlated with microvessel density. CONCLUSION CA-ADM related angiogenesis is a promising predictive marker for poor prognosis of PDAC and may provide an attractive therapeutic target for PDAC.
Collapse
|
17
|
Dai H, Jiang Y, Liu Z, Su X, Yang Y, Chen Z. Pumilio RNA-Binding Family Member 1 Plays a Promoting Role on Pancreatic Cancer Angiogenesis. Can J Gastroenterol Hepatol 2022; 2022:9202531. [PMID: 39296516 PMCID: PMC11410436 DOI: 10.1155/2022/9202531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 11/10/2022] [Accepted: 11/25/2022] [Indexed: 09/21/2024] Open
Abstract
Our previous studies showed that Pumilio RNA-binding family member 1 (PUM1) gene is abnormally expressed in pancreatic cancer (PC) tissues, and its knockdown suppresses the growth and metastasis of PC cells. Here, we aimed to further investigate its role in angiogenesis. Immunohistochemical assays were carried out to analyze CD31 and PUM1 expression levels in PC tissues and in subcutaneous xenograft tumors. CD31 levels in PC tissues are expressed as microvessel density (MVD). MVD value was positively correlated with PUM1 protein expression. PUM1 was successfully overexpressed or silenced in the PC cell lines. The proliferation, migration, invasion, and tube formation ability of HUVECs were enhanced when cocultured with PC cells overexpressing PUM1. PUM1 overexpression increased extracellular and intracellular VEGFA protein levels in PC cells. Moreover, angiogenesis-related signaling in HUVECs was activated when HUVECs were cocultured with PC cells overexpressing PUM1. Nevertheless, PC cells silenced with PUM1 had the opposite effect. Moreover, subcutaneous xenograft tumors overexpressing PUM1 have the higher expression level of CD31, while subcutaneous xenograft tumors silencing PUM1 have the lower expression level of CD31. In conclusion, PUM1 in PC cells may play a promoting role in PC angiogenesis. PUM1 may be a new regulator of angiogenesis in PC cells.
Collapse
Affiliation(s)
- Haisu Dai
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Yan Jiang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Zhipeng Liu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Xingxing Su
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Yishi Yang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Zhiyu Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| |
Collapse
|
18
|
Wu G, Ding X, Quan G, Xiong J, Li Q, Li Z, Wang Y. Hypoxia-Induced miR-210 Promotes Endothelial Cell Permeability and Angiogenesis via Exosomes in Pancreatic Ductal Adenocarcinoma. Biochem Res Int 2022; 2022:7752277. [PMID: 36466111 PMCID: PMC9718630 DOI: 10.1155/2022/7752277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 10/11/2022] [Accepted: 11/01/2022] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND Exosomes have been proven to play important diagnostic, regulatory, or communication roles in tumorigenesis, tumor progression, or metastasis; in recent studies, lots of molecules, including miRNAs, were found to be aberrantly expressed in tumor exosomes and were correlated with tumor development. However, studies about the expression, relationship, or control mechanisms of miRNAs in exosomes in pancreatic ductal adenocarcinoma (PDAC) are scarce and urgently needed. The aim of this article was to identify and investigate abnormally expressed miRNAs in PDAC exosomes in vivo and in vitro. METHODS Microarray studies were used to detect aberrantly expressed miRNAs in PDAC exosomes, and miR-210 expression in cells or exosomes was further analyzed by qRT-PCR. Bioinformatics analyses, dual-luciferase assays, WB, and other assays were utilized to explore the miRNA molecular mechanisms. The living cell coculture model and immunofluorescence analysis were employed to image the communication between PDAC cells and endothelial cells. Other biological experiments in the study include a real-time intravital imaging system, EdU, transwell, xenograft models, and so on. RESULTS miR-210 is significantly expressed in PDAC exosomes and malignant cells. High miR-210 significantly facilitated tumor angiogenesis, cell invasion, and proliferation in PDAC cells. Further mechanistic detection revealed that miR-210 negatively regulated EFNA3 expression and participated in the PI3K/AKT/VEGFA or Wnt/Β-catenin/RHOA pathways, thus promoting tumor angiogenesis and cellular permeability. PDAC cells promote endothelial angiogenesis or permeability via miR-210 transmission by tumor exosomes. Exosomal miR-210 promotes PDAC progression in vivo. Further detection of PDAC plasma exosomal miR-210 suggests that exosomal miR-210 expression was high and significantly associated with vascular invasion and TNM stage and was an independent risk factor for PDAC overall survival. CONCLUSIONS PDAC cell-secreted exosomes could promote angiogenesis and cellular permeability of neighboring endothelial angiogenesis or permeability via miR-210 transmission. Exosomal miR-210 may play important roles in tumor biology and may be a useful prognostic marker in PDAC.
Collapse
Affiliation(s)
- Guo Wu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of North Sichuan Medical College, Institute of Hepatobiliary-Pancreatic-Intestinal of North Sichuan Medical College, Nanchong, China
| | - Xiaojie Ding
- Department of Dermatology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Gang Quan
- Department of Hepatobiliary Surgery, The Affiliated Hospital of North Sichuan Medical College, Institute of Hepatobiliary-Pancreatic-Intestinal of North Sichuan Medical College, Nanchong, China
| | - Jianwei Xiong
- Department of Hepatobiliary Surgery, The Affiliated Hospital of North Sichuan Medical College, Institute of Hepatobiliary-Pancreatic-Intestinal of North Sichuan Medical College, Nanchong, China
| | - Qiang Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of North Sichuan Medical College, Institute of Hepatobiliary-Pancreatic-Intestinal of North Sichuan Medical College, Nanchong, China
| | - Zhonghu Li
- Department General Surgery, Central Theater Command General Hospital of PLA, Wuhan, China
| | - Yaqin Wang
- Department of Pathology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
19
|
Cao G, Chang Y, Yang G, Jiang Y, Han K. A novel risk score model based on four angiogenesis long non-coding RNAs for prognosis evaluation of pancreatic adenocarcinoma. Aging (Albany NY) 2022; 14:9090-9102. [PMID: 36384673 PMCID: PMC9740371 DOI: 10.18632/aging.204387] [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: 08/08/2022] [Accepted: 11/07/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) have been reported to play significant roles in tumour angiogenesis which prominently facilitates pancreatic adenocarcinoma (PAAD) progression. METHODS The clinical PAAD data were obtained from TCGA database and clinical specimens of 122 PAAD patients. The Molecular Signatures Database v4.0 was used to identify angiogenesis-related long non-coding RNAs (ARLNRs). Survival-related ARLNRs (sARLNRs) were further validated by univariate and multivariate COX regression analyses. The expressions of CASC8, AC015660.1, Z97832.2 and PAN3-AS1 in PAAD cell lines and tissues were examined by qPCR. The correlations between sARLNRs (CASC8 and AC015660.1) and clinicopathological characteristics of the 122 PAAD patients were analyzed by the chi-square test and Fisher's exact probability method. RESULTS 590 lncRNAs were identified as ARLNRs, of which four sARLNRs were further used to establish an angiogenesis-related risk score model (ARRS), by which patients in the low-risk group have better survival probabilities than those in the high-risk group. The expression levels of CASC8 and AC015660.1 were significantly higher in PAAD cell lines and tumor tissues especially in patients with advanced grades and T-stages, while Z97832.2 and PAN3-AS1 were inverse. In addition, the higher expression of CASC8 and AC015660.1 prominently associated with the larger tumour size, and the more advanced grade and T-stage. However, the relevance between the sARLNRs (CASC8 and AC015660.1) expression and lymph node metastasis status was not significant. CONCLUSIONS In the study, we illuminate the clinical significance, angiogenesis relevance and prognosis-predictive value of four sARLNRs for PAAD. The results build a bridge between sARLNRs and tumour vascularization, and also establish a reliable and accurate risk scoring model for PAAD antiangiogenic strategy.
Collapse
Affiliation(s)
- Guangbiao Cao
- Department of Hepatobiliary Surgery, Songshan General Hospital, Chongqing, China
| | - Yihang Chang
- Department of Hepatobiliary Surgery, Songshan General Hospital, Chongqing, China
| | - Guang Yang
- Department of Hepatobiliary Surgery, Songshan General Hospital, Chongqing, China
| | - Yong Jiang
- Department of Hepatobiliary Surgery, Songshan General Hospital, Chongqing, China
| | - Keqiang Han
- Department of Hepatobiliary Surgery, Songshan General Hospital, Chongqing, China
| |
Collapse
|
20
|
Jiang Z, Zhang W, Sha G, Wang D, Tang D. Galectins Are Central Mediators of Immune Escape in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:5475. [PMID: 36428567 PMCID: PMC9688059 DOI: 10.3390/cancers14225475] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers and is highly immune tolerant. Although there is immune cell infiltration in PDAC tissues, most of the immune cells do not function properly and, therefore, the prognosis of PDAC is very poor. Galectins are carbohydrate-binding proteins that are intimately involved in the proliferation and metastasis of tumor cells and, in particular, play a crucial role in the immune evasion of tumor cells. Galectins induce abnormal functions and reduce numbers of tumor-associated macrophages (TAM), natural killer cells (NK), T cells and B cells. It further promotes fibrosis of tissues surrounding PDAC, enhances local cellular metabolism, and ultimately constructs tumor immune privileged areas to induce immune evasion behavior of tumor cells. Here, we summarize the respective mechanisms of action played by different Galectins in the process of immune escape from PDAC, focusing on the mechanism of action of Galectin-1. Galectins cause imbalance between tumor immunity and anti-tumor immunity by coordinating the function and number of immune cells, which leads to the development and progression of PDAC.
Collapse
Affiliation(s)
- Zhengting Jiang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Wenjie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Gengyu Sha
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Daorong Wang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou 225000, China
| | - Dong Tang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou 225000, China
| |
Collapse
|
21
|
Ge W, Shentu D, Wang Y, Wang Y, Xue S, Yue M, Mao T, Zhang X, Xu H, Li S, Ma J, Yao J, Cui J, Wang L. A novel angiogenesis-based molecular signature related to prognosis and tumor immune interactions of pancreatic cancer. Front Cell Dev Biol 2022; 10:1001606. [PMID: 36274838 PMCID: PMC9582445 DOI: 10.3389/fcell.2022.1001606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
Angiogenesis, a hallmark of cancer, is related to prognosis, tumor progression, and treatment response. Nevertheless, the correlation of angiogenesis-based molecular signature with clinical outcome and immune cell infiltration has not been thoroughly studied in pancreatic cancer. In this study, multiple bioinformatics methods were combined to evaluate prognosis, immune cell infiltration, and the alterations of angiogenesis-related genes (ARGs) in PC samples, and further establish a novel angiogenesis-related gene signature. Moreover, the protein and mRNA expression levels of four angiogenesis risk genes were determined by Human Protein Atlas (HPA) database and qPCR analysis, respectively. Here, we recognized two distinct angiogenesis subtypes and two gene subtypes, and revealed the critical roles of ARGs in the tumor immune microenvironment (TIME), clinical features, and prognosis. Consequently, we established an ARGs score to predict prognosis and therapeutic response of PC patients, and validated its robust predictive ability. Additionally, the ARGs score was markedly associated with clinical outcomes, tumor mutation burden (TMB), and chemotherapeutic drug sensitivity. In brief, our findings imply that the ARGs score is a robust prognostic indicator and may contribute to the development of effective individualized therapies for PC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Jiujie Cui
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liwei Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
22
|
Liu T, Cheng S, Xu Q, Wang Z. Management of Advanced Pancreatic Cancer through Stromal Depletion and Immune Modulation. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58091298. [PMID: 36143975 PMCID: PMC9502806 DOI: 10.3390/medicina58091298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/10/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022]
Abstract
Pancreatic cancer is one of the leading causes of cancer-related deaths worldwide. Unfortunately, therapeutic gains in the treatment of other cancers have not successfully translated to pancreatic cancer treatments. Management of pancreatic cancer is difficult due to the lack of effective therapies and the rapid development of drug resistance. The cytotoxic agent gemcitabine has historically been the first-line treatment, but combinations of other immunomodulating and stroma-depleting drugs are currently undergoing clinical testing. Moreover, the treatment of pancreatic cancer is complicated by its heterogeneity: analysis of genomic alterations and expression patterns has led to the definition of multiple subtypes, but their usefulness in the clinical setting is limited by inter-tumoral and inter-personal variability. In addition, various cell types in the tumor microenvironment exert immunosuppressive effects that worsen prognosis. In this review, we discuss current perceptions of molecular features and the tumor microenvironment in pancreatic cancer, and we summarize emerging drug options that can complement traditional chemotherapies.
Collapse
Affiliation(s)
- Tiantong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100006, China
| | - Sihang Cheng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100006, China
| | - Qiang Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100006, China
- Correspondence: (Q.X.); (Z.W.); Tel.: +86-10-69156007 (Q.X.); +86-10-69159567 (Z.W.)
| | - Zhiwei Wang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100006, China
- Correspondence: (Q.X.); (Z.W.); Tel.: +86-10-69156007 (Q.X.); +86-10-69159567 (Z.W.)
| |
Collapse
|
23
|
Obesity and cancer-extracellular matrix, angiogenesis, and adrenergic signaling as unusual suspects linking the two diseases. Cancer Metastasis Rev 2022; 41:517-547. [PMID: 36074318 PMCID: PMC9470659 DOI: 10.1007/s10555-022-10058-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/29/2022] [Indexed: 12/24/2022]
Abstract
Obesity is an established risk factor for several human cancers. Given the association between excess body weight and cancer, the increasing rates of obesity worldwide are worrisome. A variety of obesity-related factors has been implicated in cancer initiation, progression, and response to therapy. These factors include circulating nutritional factors, hormones, and cytokines, causing hyperinsulinemia, inflammation, and adipose tissue dysfunction. The impact of these conditions on cancer development and progression has been the focus of extensive literature. In this review, we concentrate on processes that can link obesity and cancer, and which provide a novel perspective: extracellular matrix remodeling, angiogenesis, and adrenergic signaling. We describe molecular mechanisms involved in these processes, which represent putative targets for intervention. Liver, pancreas, and breast cancers were chosen as exemplary disease models. In view of the expanding epidemic of obesity, a better understanding of the tumorigenic process in obese individuals might lead to more effective treatments and preventive measures.
Collapse
|
24
|
Raja Arul GL, Toruner MD, Gatenby RA, Carr RM. Ecoevolutionary biology of pancreatic ductal adenocarcinoma. Pancreatology 2022; 22:730-740. [PMID: 35821188 DOI: 10.1016/j.pan.2022.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/01/2022] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common histological subtype of pancreatic cancer, is an aggressive disease predicted to be the 2nd cause of cancer mortality in the US by 2040. While first-line therapy has improved, 5-year overall survival has only increased from 5 to ∼10%, and surgical resection is only available for ∼20% of patients as most present with advanced disease, which is invariably lethal. PDAC has well-established highly recurrent mutations in four driver genes including KRAS, TP53, CDKN2A, and SMAD4. Unfortunately, these genetic drivers are not currently therapeutically actionable. Despite extensive sequencing efforts, few additional significantly recurrent and druggable drivers have been identified. In the absence of targetable mutations, chemotherapy remains the mainstay of treatment for most patients. Further, the role of the above driver mutations on PDAC initiation and early development is well-established. However, these mutations alone cannot account for PDAC heterogeneity nor discern early from advanced disease. Taken together, management of PDAC is an example highlighting the shortcomings of the current precision medicine paradigm. PDAC, like other malignancies, represents an ecoevolutionary process. Better understanding the disease through this lens can facilitate the development of novel therapeutic strategies to better control and cure PDAC. This review aims to integrate the current understanding of PDAC pathobiology into an ecoevolutionary framework.
Collapse
Affiliation(s)
| | - Merih D Toruner
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Robert A Gatenby
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Ryan M Carr
- Department of Oncology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
25
|
Lv F, Li X, Wang Y. Lycorine inhibits angiogenesis by docking to PDGFRα. BMC Cancer 2022; 22:873. [PMID: 35948939 PMCID: PMC9364594 DOI: 10.1186/s12885-022-09929-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 07/22/2022] [Indexed: 11/29/2022] Open
Abstract
Lycorine (Lyc) is a natural alkaloid derived from medicinal plants of the Amaryllidaceae family. Lyc has been reported to inhibit the recurrence and metastasis of different kinds of tumors. However, Lyc’s effect on angiogenesis and its specific mechanism are still not clear. This study was designed to test the antiangiogenesis effect of Lyc and to explore the possible mechanisms. We performed cell experiments to confirm Lyc’s inhibitory effect on angiogenesis and employed sunitinib as a positive control. Moreover, the synergistic effect of Lyc and sunitinib was also explored. Next, we conducted bioinformatics analyses to predict the potential targets of Lyc and verified them by western blotting and immunofluorescence. Molecular docking, kinase activity assays, Biacore assays and cellular thermal shift assays (CETSAs) were applied to elucidate the mechanism by which Lyc inhibited target activity. Lyc inhibited angiogenesis in human umbilical vein endothelial cells (HUVECs). Employing bioinformatics, we found that Lyc’s target was PDGFRα and that Lyc attenuated PDGFRα phosphorylation. We also found that Lyc inhibited PDGFRα activation by docking to it to restrain its activity. Additionally, Lyc significantly inhibited PDGF-AA-induced angiogenesis. This study provides new insights into the molecular functions of Lyc and indicates its potential as a therapeutic agent for tumor angiogenesis.
Collapse
Affiliation(s)
- Fei Lv
- Department of Oncology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110000, Liaoning Province, China
| | - XiaoQi Li
- Department of Oncology III, People's Hospital of Liaoning Provinve, Shenyang, , Liaoning, China
| | - Ying Wang
- Department of Oncology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110000, Liaoning Province, China.
| |
Collapse
|
26
|
Santos AJCA, Araújo VFS, França RPM, Silva ELA, Almeida WA, Procópio TF, Paiva PMG, Napoleão TH, Costa EVL, Nogueira RA, Pontual EV. Schinus terebinthifolia Raddi leaf lectin is an antiangiogenic agent for Coturnix japonica embryos. AN ACAD BRAS CIENC 2022; 94:e20211297. [PMID: 35920491 DOI: 10.1590/0001-3765202220211297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 11/11/2021] [Indexed: 11/22/2022] Open
Abstract
Angiogenesis (budding of new blood vessels) is involved in several processes, including the development of embryos and growth of tumors. Schinus terebinthifolia leaves express an antitumor lectin (SteLL). This work hypothesized that SteLL can interfere with the formation of a vascular network from preexisting vessels. To test this hypothesis, the effect of SteLL on the angiogenesis process was assessed using an in vivo model of yolk sac membrane of Coturnix japonica embryos. SteLL was isolated with purification factor of 46.6. As expected, polyacrylamide gel electrophoresis (PAGE) for native basic proteins confirmed the homogeneity and PAGE in presence of dodecyl sodium sulphate revealed a single 14-kDa polypeptide band. The fractal analysis by box counting and information dimension measurements indicated that SteLL at 1.35 mg/mL significantly decreased by ca. 12% the angiogenesis within the C. japonica yolk sac membrane regarding the control. The inhibition of the vascular network formation in the yolk sac membrane resulted in decreased blood supply to the embryos. Consequently, the area of embryos was significantly reduced by 9.2% regarding the control, which corroborated with the antiangiogenic activity of SteLL. The findings implicate SteLL as an antiangiogenic agent and add to the panel of biological activities of this lectin.
Collapse
Affiliation(s)
- Ardilles J C A Santos
- Universidade Federal Rural de Pernambuco, Departamento de Morfologia e Fisiologia Animal, Rua Dom Manuel de Medeiros, s/n, Dois Irmãos, 52171-300 Recife, PE, Brazil.,Universidade Federal do Piauí, Departamento de Nutrição, Rua Cícero Eduardo, s/n, Junco, 64600-000 Picos, PI, Brazil
| | - Victor F S Araújo
- Universidade Federal Rural de Pernambuco, Departamento de Morfologia e Fisiologia Animal, Rua Dom Manuel de Medeiros, s/n, Dois Irmãos, 52171-300 Recife, PE, Brazil
| | - Rayssa P M França
- Universidade Federal Rural de Pernambuco, Departamento de Morfologia e Fisiologia Animal, Rua Dom Manuel de Medeiros, s/n, Dois Irmãos, 52171-300 Recife, PE, Brazil
| | - Eva L A Silva
- Universidade Federal Rural de Pernambuco, Departamento de Morfologia e Fisiologia Animal, Rua Dom Manuel de Medeiros, s/n, Dois Irmãos, 52171-300 Recife, PE, Brazil
| | - Welton A Almeida
- Universidade Federal Rural de Pernambuco, Departamento de Morfologia e Fisiologia Animal, Rua Dom Manuel de Medeiros, s/n, Dois Irmãos, 52171-300 Recife, PE, Brazil
| | - Thamara F Procópio
- Universidade Federal de Pernambuco, Centro de Biociências, Departamento de Bioquímica, Avenida Professor Moraes Rego, s/n, Cidade Universitária, 50670-420, Recife, PE, Brazil
| | - Patrícia M G Paiva
- Universidade Federal de Pernambuco, Centro de Biociências, Departamento de Bioquímica, Avenida Professor Moraes Rego, s/n, Cidade Universitária, 50670-420, Recife, PE, Brazil
| | - Thiago H Napoleão
- Universidade Federal de Pernambuco, Centro de Biociências, Departamento de Bioquímica, Avenida Professor Moraes Rego, s/n, Cidade Universitária, 50670-420, Recife, PE, Brazil
| | - Edbhergue V L Costa
- Universidade Federal Rural de Pernambuco, Departamento de Morfologia e Fisiologia Animal, Rua Dom Manuel de Medeiros, s/n, Dois Irmãos, 52171-300 Recife, PE, Brazil
| | - Romildo A Nogueira
- Universidade Federal Rural de Pernambuco, Departamento de Morfologia e Fisiologia Animal, Rua Dom Manuel de Medeiros, s/n, Dois Irmãos, 52171-300 Recife, PE, Brazil
| | - Emmanuel V Pontual
- Universidade Federal Rural de Pernambuco, Departamento de Morfologia e Fisiologia Animal, Rua Dom Manuel de Medeiros, s/n, Dois Irmãos, 52171-300 Recife, PE, Brazil
| |
Collapse
|
27
|
Chen H, Tu W, Lu Y, Zhang Y, Xu Y, Chen X, Zhu M, Liu Y. Low-dose X-ray irradiation combined with FAK inhibitors improves the immune microenvironment and confers sensitivity to radiotherapy in pancreatic cancer. Biomed Pharmacother 2022; 151:113114. [PMID: 35594704 DOI: 10.1016/j.biopha.2022.113114] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 11/30/2022] Open
Abstract
Radiation therapy offers limited clinical benefits for patients with pancreatic cancer, partly as a result of the predominantly immunosuppressive microenvironment characteristic of this specific type of cancer. A large number of abnormal blood vessels and high-density fibrous matrices in pancreatic cancer will lead to hypoxia within tumor tissue and hinder immune cell infiltration. We used low-dose X-ray irradiation, also known as low-dose radiation therapy (LDRT), to normalize the blood vessels in pancreatic cancer, while simultaneously administering an inhibitor of focal adhesion kinase (FAK) to reduce pancreatic cancer fibrosis. We found that this treatment successfully reduced pancreatic cancer hypoxia, increased immune cell infiltration, and increased sensitivity to radiation therapy for pancreatic cancer.
Collapse
Affiliation(s)
- Huanliang Chen
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Wenzhi Tu
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Yue Lu
- Department of Radiotherapy, Huangpu Branch of the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200002, China
| | - Yingzi Zhang
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Yiqing Xu
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Xuming Chen
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Meiling Zhu
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Yong Liu
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China.
| |
Collapse
|
28
|
Gulla A, Hashimoto D, Wagner D, Damaseviciute R, Strupas K, Satoi S. Interdisciplinary Approach of Establishing PDAC Resectability: Biochemical, Radiological and NAT Regimen Prognostic Factors-Literature Review. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:756. [PMID: 35744019 PMCID: PMC9227260 DOI: 10.3390/medicina58060756] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 12/24/2022]
Abstract
Background and Objectives: Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal tumors, with a 5-year overall survival rate of less than 10%. To date, curative surgical resection remains the only favorable option for improving patients' survival. However, there is no consensus on which prognostic biochemical, radiological markers or neoadjuvant therapy regimens would benefit patients the most. Materials and Methods: A literature review was performed focusing on overall survival, R0 resection, 30-day mortality, adverse events (AEs), and elevated biomarkers. The electronic databases were searched from 2015 to 2020. Results: We reviewed 22 independent studies. In total, 20 studies were retrospective single- or multi-center reviews, while 2 studies were prospective Phase II trials. Conclusions: Patients with borderline resectable or locally advanced PDAC, who received neoadjuvant therapy (NAT) and surgery, have significantly better survival rates. The CA 19-9 biomarker levels in the neoadjuvant setting should be evaluated and considered as a specific biomarker for tumor resectability and overall survival.
Collapse
Affiliation(s)
- Aiste Gulla
- Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania; (R.D.); (K.S.)
- Department of Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius University Hospital Santaros klinikos, 08410 Vilnius, Lithuania
- Department of Surgery, Georgetown University Hospital, Washington, DC 20007, USA
| | - Daisuke Hashimoto
- Department of Surgery, Kansai Medical University, Hirakata 573-1191, Japan; (D.H.); (S.S.)
| | - Doris Wagner
- Department of Visceral, General and Transplantation Surgery, University of Graz, 3, 8010 Graz, Austria;
| | - Ryte Damaseviciute
- Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania; (R.D.); (K.S.)
| | - Kestutis Strupas
- Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania; (R.D.); (K.S.)
- Department of Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius University Hospital Santaros klinikos, 08410 Vilnius, Lithuania
| | - Sohei Satoi
- Department of Surgery, Kansai Medical University, Hirakata 573-1191, Japan; (D.H.); (S.S.)
| |
Collapse
|
29
|
Ueda G, Matsuo Y, Murase H, Aoyama Y, Kato T, Omi K, Hayashi Y, Imafuji H, Saito K, Tsuboi K, Morimoto M, Ogawa R, Takahashi H, Mitsui A, Kimura M, Takiguchi S. 10Z‑Hymenialdisine inhibits angiogenesis by suppressing NF‑κB activation in pancreatic cancer cell lines. Oncol Rep 2022; 47:48. [PMID: 35014682 PMCID: PMC8771196 DOI: 10.3892/or.2022.8259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/08/2021] [Indexed: 12/24/2022] Open
Abstract
10Z‑Hymenialdisine is a natural product derived from the marine sponge Axinella carteri. 10Z‑Hymenialdisine has anti‑inflammatory effects exerted through NF‑κB; however, it is unclear whether 10Z‑Hymenialdisine has anti‑angiogenic effects in cancer cells. In the present study, both the anti‑angiogenic and antimetastatic effects of this compound in pancreatic cancer were investigated. It was initially confirmed that 10Z‑Hymenialdisine significantly inhibited the proliferation of pancreatic cancer cells. Next, using both reverse transcription‑quantitative PCR and ELISA, it was demonstrated that 10Z‑Hymenialdisine significantly suppressed the expression of VEGF and IL‑8 mRNAs and proteins in pancreatic cancer. Immunohistochemical analysis revealed that 10Z‑Hymenialdisine inhibited NF‑κB activity in pancreatic cancer cell lines. It was also identified that 10Z‑Hymenialdisine inhibited tube formation in EA.hy926 cells. In vivo, 10Z‑Hymenialdisine significantly inhibited the growth of BxPC‑3 pancreatic cancer cells that were subcutaneously injected into model mice. In conclusion, the present study demonstrated that 10Z‑Hymenialdisine exerted anti‑angiogenic effects by suppressing NF‑κB activity and angiogenic factors, such as VEGF and IL‑8, in pancreatic cancer cell lines. 10Z‑Hymenialdisine has potential applications as a novel therapeutic agent for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Goro Ueda
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Hiromichi Murase
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Yoshinaga Aoyama
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Tomokatsu Kato
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Kan Omi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Yuichi Hayashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Hiroyuki Imafuji
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Kenta Saito
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Ken Tsuboi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
- Department of Gastrointestinal Surgery, Nagoya City East Medical Center, Chikusa-ku, Nagoya, Aichi 464-8547, Japan
| | - Mamoru Morimoto
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Ryo Ogawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Akira Mitsui
- Department of Gastroenterological Surgery, Nagoya City University West Medical Center, Kita-ku, Nagoya, Aichi 462-8508, Japan
| | - Masahiro Kimura
- Department of Gastrointestinal Surgery, Nagoya City East Medical Center, Chikusa-ku, Nagoya, Aichi 464-8547, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| |
Collapse
|
30
|
Fractal analysis improves tumour size measurement on computed tomography in pancreatic ductal adenocarcinoma: comparison with gross pathology and multi-parametric MRI. Eur Radiol 2022; 32:5053-5063. [PMID: 35201407 PMCID: PMC9279218 DOI: 10.1007/s00330-022-08631-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 12/06/2021] [Accepted: 01/31/2022] [Indexed: 11/30/2022]
Abstract
Objectives Tumour size measurement is pivotal for staging and stratifying patients with pancreatic ductal adenocarcinoma (PDA). However, computed tomography (CT) frequently underestimates tumour size due to insufficient depiction of the tumour rim. CT-derived fractal dimension (FD) maps might help to visualise perfusion chaos, thus allowing more realistic size measurement. Methods In 46 patients with histology-proven PDA, we compared tumour size measurements in routine multiphasic CT scans, CT-derived FD maps, multi-parametric magnetic resonance imaging (mpMRI), and, where available, gross pathology of resected specimens. Gross pathology was available as reference for diameter measurement in a discovery cohort of 10 patients. The remaining 36 patients constituted a separate validation cohort with mpMRI as reference for diameter and volume. Results Median RECIST diameter of all included tumours was 40 mm (range: 18–82 mm). In the discovery cohort, we found significant (p = 0.03) underestimation of tumour diameter on CT compared with gross pathology (Δdiameter3D = −5.7 mm), while realistic diameter measurements were obtained from FD maps (Δdiameter3D = 0.6 mm) and mpMRI (Δdiameter3D = −0.9 mm), with excellent correlation between the two (R2 = 0.88). In the validation cohort, CT also systematically underestimated tumour size in comparison to mpMRI (Δdiameter3D = −10.6 mm, Δvolume = −10.2 mL), especially in larger tumours. In contrast, FD map measurements agreed excellently with mpMRI (Δdiameter3D = +1.5 mm, Δvolume = −0.6 mL). Quantitative perfusion chaos was significantly (p = 0.001) higher in the tumour rim (FDrim = 4.43) compared to the core (FDcore = 4.37) and remote pancreas (FDpancreas = 4.28). Conclusions In PDA, fractal analysis visualises perfusion chaos in the tumour rim and improves size measurement on CT in comparison to gross pathology and mpMRI, thus compensating for size underestimation from routine CT. Key Points • CT-based measurement of tumour size in pancreatic adenocarcinoma systematically underestimates both tumour diameter (Δdiameter = −10.6 mm) and volume (Δvolume = −10.2 mL), especially in larger tumours. • Fractal analysis provides maps of the fractal dimension (FD), which enable a more reliable and size-independent measurement using gross pathology or multi-parametric MRI as reference standards. • FD quantifies perfusion chaos—the underlying pathophysiological principle—and can separate the more chaotic tumour rim from the tumour core and adjacent non-tumourous pancreas tissue.
Collapse
|
31
|
Cai D, Chen C, Su Y, Tan Y, Lin X, Xing R. LRG1 in pancreatic cancer cells promotes inflammatory factor synthesis and the angiogenesis of HUVECs by activating VEGFR signaling. J Gastrointest Oncol 2022; 13:400-412. [PMID: 35284128 PMCID: PMC8899736 DOI: 10.21037/jgo-21-910] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/30/2022] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND This study aimed to investigate the roles of leucine-rich alpha-2-glycoprotein 1 (LRG1) in regulating angiogenesis during pancreatic cancer (PC) pathogenesis. METHODS LRG1 expression in tissues was detected by qRT-PCR and immunohistochemistry. LRG1 in BxPC-3 and Capan-2 cells was knocked down or overexpressed. Cell viability and the migration and invasion abilities of cells were analyzed using the Cell Counting Kit-8 (CCK-8) assay and Transwell system, respectively. Interleukin-1 beta (IL-1β), IL-18, and vascular endothelial growth factor A (VEGFA) contents in cell culture were measured by ELISA, and the angiogenesis of HUVECs was assessed by the in vitro tube formation assay. In vitro LRG1 expression in BxPC-3 and Capan-2 cells was determined using immunofluorescence. RESULTS The results showed that LRG1 expression was significantly increased in pancreatic cancer tissues and cell lines. LRG1 knockdown inhibited the viability, migration, invasion, and IL-1β and IL-18 synthesis of BxPC-3 and Capan-2 cells. VEGFA synthesis in BxPC-3 and Capan-2 cells was also inhibited by LRG1 knockdown, which caused impaired tube formation of co-cultured HUVECs. LRG1 overexpression enhanced the viability, migration, and invasion of BxPC-3 and Capan-2 cells, also causing elevated tube formation of HUVECs and IL-1β and IL-18 synthesis in co-cultures of HUVECs and BxPC-3 or Capan-2 cells. Silencing of VEGF receptor (VEGFR) abrogated the enhanced tube formation and IL-1β and IL-18 synthesis in HUVECs co-cultured with BxPC-3 or Capan-2 cells overexpressing LRG1. CONCLUSIONS In conclusion, LRG1, which is highly expressed in pancreatic cancer cells, promotes inflammatory factor synthesis and the angiogenesis of HUVECs though activating the VEGFR signaling pathway.
Collapse
Affiliation(s)
- Duxiong Cai
- Department of Gastroenterology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Chunji Chen
- Department of Pathology, Hainan Provincial People’s Hospital, Haikou, China
| | - Yexiong Su
- Department of Gastroenterology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yan Tan
- Department of Gastroenterology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xuyue Lin
- Department of Gastroenterology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Rong Xing
- Department of Gastroenterology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
32
|
Role of Nrf2 in Pancreatic Cancer. Antioxidants (Basel) 2021; 11:antiox11010098. [PMID: 35052602 PMCID: PMC8773052 DOI: 10.3390/antiox11010098] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/26/2021] [Accepted: 12/28/2021] [Indexed: 12/18/2022] Open
Abstract
Pancreatic tumors are a serious health problem with a 7% mortality rate worldwide. Inflammatory processes and oxidative stress play important roles in the development of pancreatic diseases/cancer. To maintain homeostasis, a balance between free radicals and the antioxidant system is essential. Nuclear Factor Erythroid 2-Related Factor 2/NFE2L2 (Nrf2) and its negative regulator Kelch-Like ECH-Associated Protein 1 (Keap1) provide substantial protection against damage induced by oxidative stress, and a growing body of evidence points to the canonical and noncanonical Nrf2 signaling pathway as a pharmacological target in the treatment of pancreatic diseases. In this review, we present updated evidence on the activation of the Nrf2 signaling pathway and its importance in pancreatic cancer. Our review covers potential modulators of canonical and noncanonical pathway modulation mechanisms that may have a positive effect on the therapeutic response. Finally, we describe some interesting recent discoveries of novel treatments related to the antioxidant system for pancreatic cancer, including natural or synthetic compounds with therapeutic properties.
Collapse
|
33
|
Malinova A, Veghini L, Real FX, Corbo V. Cell Lineage Infidelity in PDAC Progression and Therapy Resistance. Front Cell Dev Biol 2021; 9:795251. [PMID: 34926472 PMCID: PMC8675127 DOI: 10.3389/fcell.2021.795251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/15/2021] [Indexed: 12/23/2022] Open
Abstract
Infidelity to cell fate occurs when differentiated cells lose their original identity and either revert to a more multipotent state or transdifferentiate into a different cell type, either within the same embryonic lineage or in an entirely different one. Whilst in certain circumstances, such as in wound repair, this process is beneficial, it can be hijacked by cancer cells to drive disease initiation and progression. Cell phenotype switching has been shown to also serve as a mechanism of drug resistance in some epithelial cancers. In pancreatic ductal adenocarcinoma (PDAC), the role of lineage infidelity and phenotype switching is still unclear. Two consensus molecular subtypes of PDAC have been proposed that mainly reflect the existence of cell lineages with different degrees of fidelity to pancreatic endodermal precursors. Indeed, the classical subtype of PDAC is characterised by the expression of endodermal lineage specifying transcription factors, while the more aggressive basal-like/squamous subtype is defined by epigenetic downregulation of endodermal genes and alterations in chromatin modifiers. Here, we summarise the current knowledge of mechanisms (genetic and epigenetic) of cell fate switching in PDAC and discuss how pancreatic organoids might help increase our understanding of both cell-intrinsic and cell-extrinsic factors governing lineage infidelity during the distinct phases of PDAC evolution.
Collapse
Affiliation(s)
- Antonia Malinova
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Lisa Veghini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Francisco X. Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre, Madrid, Spain
- CIBERONC, Madrid, Spain
- Department de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Vincenzo Corbo
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- ARC-Net Research Centre, University of Verona, Verona, Italy
| |
Collapse
|
34
|
Edwards P, Kang BW, Chau I. Targeting the Stroma in the Management of Pancreatic Cancer. Front Oncol 2021; 11:691185. [PMID: 34336679 PMCID: PMC8316993 DOI: 10.3389/fonc.2021.691185] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PC) presents extremely aggressive tumours and is associated with poor survival. This is attributed to the unique features of the tumour microenvironment (TME), which is known to create a dense stromal formation and poorly immunogenic condition. In particular, the TME of PC, including the stromal cells and extracellular matrix, plays an essential role in the progression and chemoresistance of PC. Consequently, several promising agents that target key components of the stroma have already been developed and are currently in multiple stages of clinical trials. Therefore, the authors review the latest available evidence on novel stroma-targeting approaches, highlighting the potential impact of the stroma as a key component of the TME in PC.
Collapse
Affiliation(s)
- Penelope Edwards
- Department of Medicine, Royal Marsden Hospital, London, United Kingdom
| | - Byung Woog Kang
- Department of Oncology/Hematology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Ian Chau
- Department of Medicine, Royal Marsden Hospital, London, United Kingdom
| |
Collapse
|
35
|
Chen J, Deng S, Zhang Y, Wang C, Hu X, Kong D, Liang G, Yuan X, Li Y, Wang X. Apatinib enhances the anti-tumor effect of paclitaxel via the PI3K/p65/Bcl-xl pathway in triple-negative breast cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1001. [PMID: 34277801 PMCID: PMC8267319 DOI: 10.21037/atm-21-805] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 06/04/2021] [Indexed: 12/14/2022]
Abstract
Background Apatinib is a new generation of small molecule tyrosine kinase inhibitor, which can highly selectively inhibit phosphorylation of vascular endothelial growth factor receptor 2 (VEGFR-2). This study aimed to investigate the synergistic effects of apatinib and paclitaxel (PTX) on triple-negative breast cancer (TNBC) in vivo and in vitro, and to explore the molecular mechanism of the PI3K/p65/Bcl-xl pathway. Methods In vitro, 3-(4,5-dimethylthiazol-2-Yl)-2,5-diphenyltetrazolium bromide (MTT) method, flow cytometry (FCM), wound healing assay, and transwell matrix assay were conducted to measure the effects of apatinib and PTX on cell viability, apoptosis, migration, and invasion in TNBC cell line MDA-MB-468. Western blot (WB) was conducted to detect protein expression levels of PI3K, p65, and Bcl-xl after the application of apatinib and PTX. In vivo, MDA-MB-468 tumor-bearing nude mice were treated with apatinib and PTX, and tumor growth was observed. Results In vitro, apatinib and PTX could synergistically suppress the cell viability, the combined group had the most obvious effect. Apatinib and PTX could promote apoptosis and suppress migration and invasion of TNBC cells. Apatinib could reduce the expression of p-PI3K, p65, and Bcl-xl proteins (P<0.05). In vivo, apatinib and PTX could inhibit tumor size and weight of model mice, and the combined agents had a more significant effect. Conclusions Apatinib could enhance the anti-tumor effect of PTX on TNBC cells through the PI3K/p65/Bcl-xl molecular pathway, and apatinib combined with PTX might be a promising option for TNBC treatment.
Collapse
Affiliation(s)
- Jing Chen
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Shuzhen Deng
- School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Yifan Zhang
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Chaokun Wang
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Xiaochen Hu
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Dejiu Kong
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Gaofeng Liang
- School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Xiang Yuan
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, CA, USA
| | - Xinshuai Wang
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
36
|
Omi K, Matsuo Y, Ueda G, Aoyama Y, Kato T, Hayashi Y, Imafuji H, Saito K, Tsuboi K, Morimoto M, Ogawa R, Takahashi H, Takiguchi S. Escin inhibits angiogenesis by suppressing interleukin‑8 and vascular endothelial growth factor production by blocking nuclear factor‑κB activation in pancreatic cancer cell lines. Oncol Rep 2021; 45:55. [PMID: 33760162 PMCID: PMC7962110 DOI: 10.3892/or.2021.8006] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 12/21/2020] [Indexed: 12/20/2022] Open
Abstract
Pancreatic cancer (PaCa) is one of the most aggressive types of cancer. Thus, the development of new and more effective therapies is urgently required. Escin, a pentacyclic triterpenoid from the horse chestnut, has been reported to exhibit antitumor potential by reducing cell proliferation and blocking the nuclear factor‑κB (NF‑κB) signaling pathway in several types of cancer. Our previous study reported that NF‑κB enhanced the secretion of interleukin (IL)‑8 and vascular endothelial growth factor (VEGF), thereby inducing angiogenesis in PaCa cell lines. In the present study, it was examined whether escin inhibited angiogenesis by blocking NF‑κB activation in PaCa. It was initially confirmed that escin, at concentrations >10 µM, significantly inhibited the proliferation of several PaCa cell lines. Next, using immunocytochemical staining, it was found that escin inhibited the nuclear translocation of NF‑κB. Furthermore, ELISA confirmed that NF‑κB activity in the escin‑treated PaCa cells was significantly inhibited and reverse transcription‑quantitative PCR showed that the mRNA expression levels of tumor necrosis factor‑α‑induced IL‑8 and VEGF were significantly suppressed following escin treatment in the PaCa cell lines. ELISA also showed that escin decreased the secretion of IL‑8 and VEGF from the PaCa cells. Furthermore, tube formation in immortalized human endothelial cells was inhibited following incubation with the supernatants from escin‑treated PaCa cells. These results indicated that escin inhibited angiogenesis by reducing the secretion of IL‑8 and VEGF by blocking NF‑κB activity in PaCa. In conclusion, escin could be used as a novel molecular therapy for PaCa.
Collapse
Affiliation(s)
- Kan Omi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Goro Ueda
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Yoshinaga Aoyama
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Tomokatsu Kato
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Yuichi Hayashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Hiroyuki Imafuji
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Kenta Saito
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Ken Tsuboi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Mamoru Morimoto
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Ryo Ogawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| |
Collapse
|
37
|
Zhang X, Liu Y, Zhang Z, Tan J, Zhang J, Ou H, Li J, Song Z. Multi-Omics Analysis of Anlotinib in Pancreatic Cancer and Development of an Anlotinib-Related Prognostic Signature. Front Cell Dev Biol 2021; 9:649265. [PMID: 33748143 PMCID: PMC7969999 DOI: 10.3389/fcell.2021.649265] [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: 01/04/2021] [Accepted: 02/08/2021] [Indexed: 12/28/2022] Open
Abstract
Aberrant regulation of angiogenesis involves in the growth and metastasis of tumors, but angiogenesis inhibitors fail to improve overall survival of pancreatic cancer patients in previous phase III clinical trials. A comprehensive knowledge of the mechanism of angiogenesis inhibitors against pancreatic cancer is helpful for clinical purpose and for the selection of patients who might benefit from the inhibitors. In this work, multi-omics analyses (transcriptomics, proteomics, and phosphoproteomics profiling) were carried out to delineate the mechanism of anlotinib, a novel angiogenesis inhibitor, against pancreatic cancer cells. The results showed that anlotinib exerted noteworthy cytotoxicity on pancreatic cancer cells. Multi-omics analyses revealed that anlotinib had a profound inhibitory effect on ribosome, and regulated cell cycle, RNA metabolism and lysosome. Based on the multi-omics results and available data deposited in public databases, an anlotinib-related gene signature was further constructed to identify a subgroup of pancreatic cancer patients who had a dismal prognosis and might be responsive to anlotinib.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yang Liu
- Department of Pathology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhen Zhang
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Juan Tan
- Department of Pathology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Junjun Zhang
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Hao Ou
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jie Li
- Department of Information Science and Engineering, Hunan University of Chinese Medicine, Changsha, China
| | - Zewen Song
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
38
|
Chen K, Wang Q, Kornmann M, Tian X, Yang Y. The Role of Exosomes in Pancreatic Cancer From Bench to Clinical Application: An Updated Review. Front Oncol 2021; 11:644358. [PMID: 33718244 PMCID: PMC7952979 DOI: 10.3389/fonc.2021.644358] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 01/12/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most dismal gastrointestinal malignancies with an overall 5-year survival rate of 8%-9%. The intra-tumor heterogeneity and special tumor microenvironment in PDAC make it challenging to develop effective treatment strategies. Exosomes are extracellular vesicles that originate from the endosomes and have a diameter of 40-160 nm. A growing body of evidence has shown that exosomes play vital roles in tumor initiation and development. Recently, extensive application of exosomes as biomarkers and drug carriers has rendered them attractive in the field of PDAC. This review summarizes the latest progress in the methodologies for isolation, modification, and tracking of exosomes, exosome-mediated cell-to-cell communication, clinical applications of exosome as minimally invasive liquid biopsy and drugs carriers, as well as their involvement in the angiogenic regulation in PDAC. In spite of these advancements, some obstacles are still required to be overcome to use the exosome-based technologies for early diagnosis or improvement of prognosis of patients with PDAC.
Collapse
Affiliation(s)
- Kai Chen
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Qi Wang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Marko Kornmann
- Clinic of General, Visceral and Transplantation Surgery, University of Ulm, Ulm, Germany
| | - Xiaodong Tian
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Yinmo Yang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
39
|
Prognostic value of Glypican family genes in early-stage pancreatic ductal adenocarcinoma after pancreaticoduodenectomy and possible mechanisms. BMC Gastroenterol 2020; 20:415. [PMID: 33302876 PMCID: PMC7731467 DOI: 10.1186/s12876-020-01560-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 11/24/2020] [Indexed: 01/05/2023] Open
Abstract
Background This study explored the prognostic significance of Glypican (GPC) family genes in patients with pancreatic ductal adenocarcinoma (PDAC) after pancreaticoduodenectomy using data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). Methods A total of 112 PDAC patients from TCGA and 48 patients from GEO were included in the analysis. The relationship between overall survival and the expression of GPC family genes as well as basic clinical characteristics was analyzed using the Kaplan-Meier method with the log-rank test. Joint effects survival analysis was performed to further examine the relationship between GPC genes and prognosis. A prognosis nomogram was established based on clinical characteristics and prognosis-related genes. Prognosis-related genes were investigated by genome-wide co-expression analysis and gene set enrichment analysis (GSEA) was carried out to identify potential mechanisms of these genes affecting prognosis. Results In TCGA database, high expression of GPC2, GPC3, and GPC5 was significantly associated with favorable survival (log-rank P = 0.031, 0.021, and 0.028, respectively; adjusted P value = 0.005, 0.022, and 0.020, respectively), and joint effects analysis of these genes was effective for prognosis prediction. The prognosis nomogram was applied to predict the survival probability using the total scores calculated. Genome-wide co-expression and GSEA analysis suggested that the GPC2 may affect prognosis through sequence-specific DNA binding, protein transport, cell differentiation and oncogenic signatures (KRAS, RAF, STK33, and VEGFA). GPC3 may be related to cell adhesion, angiogenesis, inflammatory response, signaling pathways like Ras, Rap1, PI3K-Akt, chemokine, GPCR, and signatures like cyclin D1, p53, PTEN. GPC5 may be involved in transcription factor complex, TFRC1, oncogenic signatures (HOXA9 and BMI1), gene methylation, phospholipid metabolic process, glycerophospholipid metabolism, cell cycle, and EGFR pathway. Conclusion GPC2, GPC3, and GPC5 expression may serve as prognostic indicators in PDAC, and combination of these genes showed a higher efficiency for prognosis prediction.
Collapse
|
40
|
Geismann C, Arlt A. Coming in the Air: Hypoxia Meets Epigenetics in Pancreatic Cancer. Cells 2020; 9:cells9112353. [PMID: 33113836 PMCID: PMC7694089 DOI: 10.3390/cells9112353] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/17/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023] Open
Abstract
With a five-year survival rate under 9%, pancreatic ductal adenocarcinoma (PDAC) represents one of the deadliest tumors. Although the treatment options are slightly improving, PDAC is the second leading cause of cancer related death in 2020 in the US. In addition to a pronounced desmoplastic stroma reaction, pancreatic cancer is characterized by one of the lowest levels of oxygen availability within the tumor mass and these hypoxic conditions are known to contribute to tumor development and progression. In this context, the major hypoxia associated transcription factor family, HIF, regulates hundreds of genes involved in angiogenesis, metabolism, migration, invasion, immune escape and therapy resistance. Current research implications show, that hypoxia also modulates diverse areas of epigenetic mechanisms like non-coding RNAs, histone modifications or DNA methylation, which cooperate with the hypoxia-induced transcription factors as well as directly regulate the hypoxic response pathways. In this review, we will focus on hypoxia-mediated epigenetic alterations and their impact on pancreatic cancer.
Collapse
Affiliation(s)
- Claudia Geismann
- Laboratory of Molecular Gastroenterology & Hepatology, Department of Internal Medicine I, UKSH-Campus Kiel, 24105 Kiel, Germany;
| | - Alexander Arlt
- Laboratory of Molecular Gastroenterology & Hepatology, Department of Internal Medicine I, UKSH-Campus Kiel, 24105 Kiel, Germany;
- Department for Gastroenterology, European Medical School (EMS), Klinikum Oldenburg AöR, 26133 Oldenburg, Germany
- Correspondence: ; Tel.: +49-441-403-2581
| |
Collapse
|
41
|
Ko JH, Kwon HS, Kim B, Min G, Shin C, Yang SW, Lee SW, Lee Y, Hong D, Kim YS. Preclinical Efficacy and Safety of an Anti-Human VEGFA and Anti-Human NRP1 Dual-Targeting Bispecific Antibody (IDB0076). Biomolecules 2020; 10:biom10060919. [PMID: 32560565 PMCID: PMC7356919 DOI: 10.3390/biom10060919] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/04/2020] [Accepted: 06/15/2020] [Indexed: 01/13/2023] Open
Abstract
Although bevacizumab (Avastin®) has been approved as an antiangiogenic agent against some cancers, the efficacy is transient and unsatisfactory in other cancers most likely owing to the presence of alternative proangiogenic factors. Therefore, simultaneous blocking of several proangiogenic factors may be a promising strategy for antiangiogenic cancer therapeutics. Accordingly, neuropilin-1 (NRP1) is an attractive target because it serves as a multifunctional receptor for the vascular endothelial growth factor (VEGF) family. Here, we aimed to generate and test an anti-VEGFA and anti-NRP1 dual-targeting bispecific antibody (named as IDB0076) by genetic fusion of an NRP1-targeting peptide to the C-terminus of the bevacizumab heavy chain. Similar to the parental antibody (bevacizumab), IDB0076 suppressed VEGFA-induced migration of human endothelial cells. In contrast, IDB0076 inhibited endothelial-cell migration induced by other angiogenesis growth factors and manifested a more potent antitumor activity than that of bevacizumab in a murine tumor xenograft model. When toxicity was preliminarily evaluated in cynomolgus monkeys, IDB0076 showed no substantial adverse effects, e.g., the absence of noticeable nephrotoxicity, which has previously been documented for the combination therapy of bevacizumab and an anti-NRP1 antibody. Thus, VEGFA-and-NRP1 dual-targeting bispecific antibody IDB0076 may be a potent and safe anticancer agent worthy of further preclinical and clinical studies.
Collapse
Affiliation(s)
- Jong-Hee Ko
- Research Laboratory, ILDONG Pharmaceutical Co., Ltd., Hwaseong 18449, Korea; (J.-H.K.); (H.-S.K.); (B.K.); (G.M.); (C.S.); (S.-W.Y.); (S.W.L.); (Y.L.); (D.H.)
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
| | - Hyuk-Sang Kwon
- Research Laboratory, ILDONG Pharmaceutical Co., Ltd., Hwaseong 18449, Korea; (J.-H.K.); (H.-S.K.); (B.K.); (G.M.); (C.S.); (S.-W.Y.); (S.W.L.); (Y.L.); (D.H.)
| | - Bomin Kim
- Research Laboratory, ILDONG Pharmaceutical Co., Ltd., Hwaseong 18449, Korea; (J.-H.K.); (H.-S.K.); (B.K.); (G.M.); (C.S.); (S.-W.Y.); (S.W.L.); (Y.L.); (D.H.)
| | - Gihong Min
- Research Laboratory, ILDONG Pharmaceutical Co., Ltd., Hwaseong 18449, Korea; (J.-H.K.); (H.-S.K.); (B.K.); (G.M.); (C.S.); (S.-W.Y.); (S.W.L.); (Y.L.); (D.H.)
| | - Chorong Shin
- Research Laboratory, ILDONG Pharmaceutical Co., Ltd., Hwaseong 18449, Korea; (J.-H.K.); (H.-S.K.); (B.K.); (G.M.); (C.S.); (S.-W.Y.); (S.W.L.); (Y.L.); (D.H.)
| | - Seok-Woo Yang
- Research Laboratory, ILDONG Pharmaceutical Co., Ltd., Hwaseong 18449, Korea; (J.-H.K.); (H.-S.K.); (B.K.); (G.M.); (C.S.); (S.-W.Y.); (S.W.L.); (Y.L.); (D.H.)
| | - Seong Wook Lee
- Research Laboratory, ILDONG Pharmaceutical Co., Ltd., Hwaseong 18449, Korea; (J.-H.K.); (H.-S.K.); (B.K.); (G.M.); (C.S.); (S.-W.Y.); (S.W.L.); (Y.L.); (D.H.)
| | - Youngmin Lee
- Research Laboratory, ILDONG Pharmaceutical Co., Ltd., Hwaseong 18449, Korea; (J.-H.K.); (H.-S.K.); (B.K.); (G.M.); (C.S.); (S.-W.Y.); (S.W.L.); (Y.L.); (D.H.)
| | - Dahae Hong
- Research Laboratory, ILDONG Pharmaceutical Co., Ltd., Hwaseong 18449, Korea; (J.-H.K.); (H.-S.K.); (B.K.); (G.M.); (C.S.); (S.-W.Y.); (S.W.L.); (Y.L.); (D.H.)
| | - Yong-Sung Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
- Correspondence: ; Tel.: +82 31-219-2662
| |
Collapse
|
42
|
Garcia J, Hurwitz HI, Sandler AB, Miles D, Coleman RL, Deurloo R, Chinot OL. Bevacizumab (Avastin®) in cancer treatment: A review of 15 years of clinical experience and future outlook. Cancer Treat Rev 2020; 86:102017. [PMID: 32335505 DOI: 10.1016/j.ctrv.2020.102017] [Citation(s) in RCA: 681] [Impact Index Per Article: 136.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/20/2020] [Accepted: 03/22/2020] [Indexed: 01/01/2023]
Abstract
When the VEGF-A-targeting monoclonal antibody bevacizumab (Avastin®) entered clinical practice more than 15 years ago, it was one of the first targeted therapies and the first approved angiogenesis inhibitor. Marking the beginning for a new line of anti-cancer treatments, bevacizumab remains the most extensively characterized anti-angiogenetic treatment. Initially approved for treatment of metastatic colorectal cancer in combination with chemotherapy, its indications now include metastatic breast cancer, non-small-cell lung cancer, glioblastoma, renal cell carcinoma, ovarian cancer and cervical cancer. This review provides an overview of the clinical experience and lessons learned since bevacizumab's initial approval, and highlights how this knowledge has led to the investigation of novel combination therapies. In the past 15 years, our understanding of VEGF's role in the tumor microenvironment has evolved. We now know that VEGF not only plays a major role in controlling blood vessel formation, but also modulates tumor-induced immunosuppression. These immunomodulatory properties of bevacizumab have opened up new perspectives for combination therapy approaches, which are being investigated in clinical trials. Specifically, the combination of bevacizumab with cancer immunotherapy has recently been approved in non-small-cell lung cancer and clinical benefit was also demonstrated for treatment of hepatocellular carcinoma. However, despite intense investigation, reliable and validated biomarkers that would enable a more personalized use of bevacizumab remain elusive. Overall, bevacizumab is expected to remain a key agent in cancer therapy, both due to its established efficacy in approved indications and its promise as a partner in novel targeted combination treatments.
Collapse
Affiliation(s)
- Josep Garcia
- Global Clinical Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| | | | | | | | - Robert L Coleman
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas, MD Anderson Cancer Center, TX, USA
| | - Regula Deurloo
- Oncology Biomarker Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Olivier L Chinot
- Aix-Marseille University, Assistance Publique-Hopitaux de Marseille, Centre Hospitalo-Universitaire Timone, Service de Neuro-Oncologie, Marseille, France
| |
Collapse
|
43
|
Liu X, Xu H, Chen M, Tan X, Chen X, Yang Y, Lin M, Liu G, Liang X, Qian Y, Yuan G, Chen M, Li W, Miao H, Li M, Liao X, Dai W, Chen N. Identify potential clinical significance of long noncoding RNA forkhead box P4 antisense RNA 1 in patients with early stage pancreatic ductal adenocarcinoma. Cancer Med 2020; 9:2062-2076. [PMID: 31991068 PMCID: PMC7064149 DOI: 10.1002/cam4.2818] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/04/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022] Open
Abstract
Previous studies have shown that forkhead box P4 antisense RNA 1 (FOXP4-AS1) is dysregulated in tumor tissues and can serve as a prognostic indicator for multiple cancers. However, the clinical significance of FOXP4-AS1 in pancreatic ductal adenocarcinoma (PDAC) remains unclear. The goal of this study is to recognize the possible clinical significance of long noncoding RNA FOXP4-AS1 in patients with early stage PDAC. A total of 112 patients from The Cancer Genome Atlas (TCGA) PDAC cohort, receiving RNA sequencing, were involved in the study. Survival analysis, functional mechanism, and potential small molecule drugs of target therapy of FOXP4-AS1 were performed in this study. Survival analysis in TCGA PDAC cohort suggested that patients with high FOXP4-AS1 expression had significantly augmented possibility of death than in PDAC patients with lower FOXP4-AS1 expression (adjusted P = .008; adjusted HR = 2.143, 95% CI = 1.221-3.760). In this study, a genome-wide RNA sequencing dataset was used to identify 927 genes co-expressing with FOXP4-AS1 in PDAC tumor tissues. A total of 676 differentially expressed genes were identified between different FOXP4-AS1 expression groups. Functional enrichment analysis of these genes and gene set enrichment analysis for PDAC genome-wide RNA sequencing dataset was done. We have found that FOXP4-AS1 may function in PDAC by participating in biological processes and pathways including oxidative phosphorylation, tricarboxylic acid cycle, classical tumor-related pathways such as NF-kappaB as well as Janus kinase/signal transducers in addition to activators of transcription, cell proliferation, and adhesion. In addition, we also screened two potential targeted therapeutic small molecule drugs (dimenhydrinate and metanephrine) for FOXP4-AS1 in PDAC. In conclusion, our present study demonstrated that higher expression of FOXP4-AS1 in PDAC tumor tissues were related with an inferior medical outcome. Through multiple genome-wide approaches, we identified the potential molecular mechanisms of FOXP4-AS1 in PDAC and two targeted therapeutic drugs for it.
Collapse
Affiliation(s)
- Xiao‐Guang Liu
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Hao Xu
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Ming Chen
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Xiao‐Yu Tan
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Xiao‐Feng Chen
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Yong‐Guang Yang
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Man‐Zhou Lin
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Guo‐Hua Liu
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Xiao‐Lu Liang
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Yi‐Bin Qian
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Guo‐Jia Yuan
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Min‐Qiang Chen
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Wen‐Tao Li
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Hui‐Lai Miao
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Ming‐Yi Li
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Xi‐Wen Liao
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxi Zhuang Autonomous RegionPeople's Republic of China
| | - Wei Dai
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| | - Nian‐Ping Chen
- Department of Hepatobiliary SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvincePeople's Republic of China
| |
Collapse
|
44
|
Guerin MV, Finisguerra V, Van den Eynde BJ, Bercovici N, Trautmann A. Preclinical murine tumor models: a structural and functional perspective. eLife 2020; 9:e50740. [PMID: 31990272 PMCID: PMC6986875 DOI: 10.7554/elife.50740] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/06/2020] [Indexed: 12/14/2022] Open
Abstract
The goal of this review is to pinpoint the specific features, including the weaknesses, of various tumor models, and to discuss the reasons why treatments that are efficient in murine tumor models often do not work in clinics. In a detailed comparison of transplanted and spontaneous tumor models, we focus on structure-function relationships in the tumor microenvironment. For instance, the architecture of the vascular tree, which depends on whether tumor cells have gone through epithelial-mesenchymal transition, is determinant for the extension of the spontaneous necrosis, and for the intratumoral localization of the immune infiltrate. Another key point is the model-dependent abundance of TGFβ in the tumor, which controls the variable susceptibility of different tumor models to treatments. Grounded in a historical perspective, this review provides a rationale for checking factors that will be key for the transition between preclinical murine models and clinical applications.
Collapse
Affiliation(s)
- Marion V Guerin
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, F-75014ParisFrance
| | - Veronica Finisguerra
- Ludwig Institute for Cancer Research, de Duve Institute WELBIOUCLouvainBrusselsBelgium
| | | | - Nadege Bercovici
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, F-75014ParisFrance
| | - Alain Trautmann
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, F-75014ParisFrance
| |
Collapse
|
45
|
Pan S, Hu Y, Hu M, Xu Y, Chen M, Du C, Cui J, Zheng P, Lai J, Zhang Y, Bai J, Jiang P, Zhu J, He Y, Wang J. S100A8 facilitates cholangiocarcinoma metastasis via upregulation of VEGF through TLR4/NF‑κB pathway activation. Int J Oncol 2019; 56:101-112. [PMID: 31746424 PMCID: PMC6910197 DOI: 10.3892/ijo.2019.4907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/03/2019] [Indexed: 12/12/2022] Open
Abstract
A growing body of evidence indicates that S100 calcium-binding protein A8 (S100A8) is frequently overexpressed in malignant tumor tissues and regulates tumor progression; however, the role of S100A8 in cholangiocarcinoma (CCA) remains unclear. The present study demonstrated that the protein expression of S100A8 was significantly higher in pathological tissues compared with adjacent normal tissues from patients with CCA. In addition, S100A8 expression was significantly associated with differentiation, lymph node metastasis and poor prognosis in patients following surgical resection of CCA. Furthermore, both in vitro and in vivo experiments revealed that overexpression of S100A6 promoted, while S100A8 knockdown attenuated, the migration and metastasis of CCA cells. Of note, the present results indicated that S100A8 promoted the CCA tumor cell-induced migration of vascular endothelial cells. Finally, S100A8 was demonstrated to positively regulate the expression of vascular endothelial growth factor (VEGF) in CCA cells, which was mediated by activation of the Toll-like receptor 4 (TLR4)/NF-κB pathway. In conclusion, the present study demonstrated that S100A8 had an important role in facilitating CCA cell migration and metastasis via upregulation of VEGF expression by activating the TLR4/NF-κB pathway. These findings may provide a novel target for CCA treatment.
Collapse
Affiliation(s)
- Shuguang Pan
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Centre for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Ying Hu
- Oncology Department, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Mengjia Hu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Centre for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yang Xu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Centre for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Mo Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Centre for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Changhong Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Centre for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jinchi Cui
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Ping Zheng
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jiejuan Lai
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yujun Zhang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jie Bai
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Peng Jiang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jin Zhu
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yu He
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Junping Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Centre for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
46
|
Network Meta-Analysis of Efficacy and Safety of Chemotherapy and Target Therapy in the First-Line Setting of Advanced Pancreatic Cancer. Cancers (Basel) 2019; 11:cancers11111746. [PMID: 31703359 PMCID: PMC6895788 DOI: 10.3390/cancers11111746] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/29/2019] [Accepted: 11/02/2019] [Indexed: 12/13/2022] Open
Abstract
Both gemcitabine and fluoropyrimidine are recommended backbones in the first-line treatment of pancreatic ductal adenocarcinoma (PDAC). To compare the efficacy and safety of these two therapeutic backbones, and to investigate the optimal therapies, we conducted a network meta-analysis. By retrospective analysis of randomized controlled trials (RCT), the most preferred therapeutic regimen may be predicted. The eligible RCTs of the gemcitabine-based therapies and fluoropyrimidine-based therapies were searched up to 31 August 2019. In a frequentist network meta-analysis, treatments were compared and ranked according to overall survival (OS) and progression-free survival (PFS). Thirty-two trials with 10,729 patients were included. The network meta-analyses results for overall survival and progression-free survival showed that fluoropyrimidine-based therapy seems to be the most effective treatment choice. Compared to gemcitabine combined with taxanes or immunotherapy, fluoropyrimidine-based therapy had comparable treatment effects (PFS: 0.67, p-Value = 0.11; 0.76, p-Value = 0.32; OS: 0.80, p-Value = 0.16; 0.77, p-Value = 0.21). Moreover, the combination of immunotherapy and gemcitabine had tolerable toxicities. Based on current evidence, fluoropyrimidine-based therapies and the combination of gemcitabine and taxanes were the most effective therapies in the advanced pancreatic cancer, and the combination of immunotherapy and gemcitabine can be developed into a new form of therapy.
Collapse
|
47
|
Kajtazi Y, Kaemmerer D, Sänger J, Schulz S, Lupp A. Somatostatin and chemokine CXCR4 receptor expression in pancreatic adenocarcinoma relative to pancreatic neuroendocrine tumours. J Cancer Res Clin Oncol 2019; 145:2481-2493. [PMID: 31451931 DOI: 10.1007/s00432-019-03011-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Pancreatic adenocarcinoma (PAC) represents one of the most fatal types of cancer with an exceptionally poor prognosis, underscoring the need for improved diagnostic and treatment approaches. An over-expression of somatostatin receptors (SST) as well as of the chemokine receptor CXCR4 has been shown for many tumour entities. Respective expression data for PAC, however, are scarce and contradictory. METHODS Overall, 137 tumour samples from 70 patients, 26 of whom were diagnosed with PAC and 44 with pancreatic neuroendocrine tumour (PanNET), were compared in terms of SST and CXCR4 expression by immunohistochemical analysis using well-characterized rabbit monoclonal antibodies. RESULTS Only SST1 and CXCR4 expression was detected in PAC tumours, with SST1 present in 42.3% and CXCR4 in 7.7% of cases. However, the overall staining intensity was very weak. In contrast to the tumour cells, in many PAC cases, tumour capillaries exhibited strong SST3, SST5, or CXCR4 expression. In PanNETs, SST2 was the most-prominently expressed receptor, observed in 75.0% of the tumours at medium-strong intensity. SST5, SST1, and CXCR4 expression was detected in 20.5%, 15.9%, and 11.4% of PanNET cases, respectively, but the staining intensity was only weak. SST2 positivity in PanNET, but not in PAC, was associated with favourable patient outcomes. CONCLUSIONS SST or CXCR4 expression in PAC is clearly of no therapeutic relevance. However, indirect targeting of these tumours via SST3, SST5, or CXCR4 on tumour microvessels may represent a promising additional therapeutic strategy.
Collapse
Affiliation(s)
- Ylberta Kajtazi
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Daniel Kaemmerer
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Jörg Sänger
- Laboratory of Pathology and Cytology Bad Berka, Bad Berka, Germany
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany.
| |
Collapse
|
48
|
Chen M, Yang S, Fan L, Wu L, Chen R, Chang J, Hu J. Combined Antiangiogenic Therapy and Immunotherapy Is Effective for Pancreatic Cancer With Mismatch Repair Proficiency but High Tumor Mutation Burden: A Case Report. Pancreas 2019; 48:1232-1236. [PMID: 31609933 PMCID: PMC6830947 DOI: 10.1097/mpa.0000000000001398] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/13/2019] [Indexed: 12/20/2022]
Abstract
Immunotherapy has been recommended as a second-line treatment only for high microsatellite instability or DNA mismatch repair deficiency advanced pancreatic cancer in National Comprehensive Cancer Network guidelines. Here, we report a case with a good response to immunotherapy in pancreatic cancer with mismatch repair proficiency. A 55-year-old woman diagnosed with pancreatic cancer cT4N1M1 (liver, lung) who harbored ERBB2 mutations with high tumor mutation burden (TMB) underwent multiple therapies and survived 19 months. A partial response in pancreatic cancer was observed when the patient was treated with combined antiangiogenic therapy and immunotherapy after a series of ineffective treatments. Neutrophil-to-lymphocyte ratio (NLR), a predictive marker of efficacy of immunotherapy, confirmed that immunotherapy resulted in the partial response in pancreatic cancer. To our knowledge, this is the first to report advanced pancreatic cancer with mismatch repair proficiency had a good response to immunotherapy, and this is the first to report an association between high blood-based TMB or NLR and improved clinical outcomes in pancreatic cancer. Therefore, TMB may also be a biomarker for immunotherapy of pancreatic cancer, and NLR may be a prospective predictive marker for efficacy of immunotherapy in pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | - Lu Wu
- Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | - Jianli Hu
- Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|