1
|
Yuan T, Fu X, Hu R, Zheng X, Jiang D, Jing L, Kuang X, Guo Z, Luo X, Liu Y, Zou X, Luker GD, Mi S, Liu C, Sun W. Bioprinted, spatially defined breast tumor microenvironment models of intratumoral heterogeneity and drug resistance. Trends Biotechnol 2024; 42:1523-1550. [PMID: 39112274 DOI: 10.1016/j.tibtech.2024.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 11/17/2024]
Abstract
Cellular, extracellular matrix (ECM), and spatial heterogeneity of tumor microenvironments (TMEs) regulate disease progression and treatment efficacy. Developing in vitro models that recapitulate the TME promises to accelerate studies of tumor biology and identify new targets for therapy. Here, we used extrusion-based, multi-nozzle 3D bioprinting to spatially pattern triple-negative MDA-MB-231 breast cancer cells, endothelial cells (ECs), and human mammary cancer-associated fibroblasts (HMCAFs) with biomimetic ECM inks. Bioprinted models captured key features of the spatial architecture of human breast tumors, including varying-sized dense regions of cancer cells and surrounding microvessel-rich stroma. Angiogenesis and ECM stiffening occurred in the stromal area but not the cancer cell-rich (CCR) regions, mimicking pathological changes in patient samples. Transcriptomic analyses revealed upregulation of angiogenesis-related and ECM remodeling-related signatures in the stroma region and identified potential ligand-receptor (LR) mediators of these processes. Breast cancer cells in distinct parts of the bioprinted TME showed differing sensitivities to chemotherapy, highlighting environmentally mediated drug resistance. In summary, our 3D-bioprinted tumor model will act as a platform to discover integrated functions of the TME in cancer biology and therapy.
Collapse
Affiliation(s)
- Tianying Yuan
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, 518055, Shenzhen, China; Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China
| | - Xihong Fu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Rongcheng Hu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Xiaochun Zheng
- Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China
| | - Dong Jiang
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Lanyu Jing
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Xiaying Kuang
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Zhongwei Guo
- School of Mechanics and Safety Engineering, Zhengzhou University, 450001, Zhengzhou, China
| | - Xu Luo
- Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China
| | - Yixin Liu
- Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Gary D Luker
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Shengli Mi
- Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China.
| | - Chun Liu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China.
| | - Wei Sun
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, 518055, Shenzhen, China; Department of Mechanical Engineering, Biomanufacturing Center, Tsinghua University, 100084, Beijing, China; Department of Mechanical Engineering, Drexel University, Philadelphia, PA 19104, USA.
| |
Collapse
|
2
|
Banerjee A, Farci P. Fibrosis and Hepatocarcinogenesis: Role of Gene-Environment Interactions in Liver Disease Progression. Int J Mol Sci 2024; 25:8641. [PMID: 39201329 PMCID: PMC11354981 DOI: 10.3390/ijms25168641] [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: 06/26/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
The liver is a complex organ that performs vital functions in the body. Despite its extraordinary regenerative capacity compared to other organs, exposure to chemical, infectious, metabolic and immunologic insults and toxins renders the liver vulnerable to inflammation, degeneration and fibrosis. Abnormal wound healing response mediated by aberrant signaling pathways causes chronic activation of hepatic stellate cells (HSCs) and excessive accumulation of extracellular matrix (ECM), leading to hepatic fibrosis and cirrhosis. Fibrosis plays a key role in liver carcinogenesis. Once thought to be irreversible, recent clinical studies show that hepatic fibrosis can be reversed, even in the advanced stage. Experimental evidence shows that removal of the insult or injury can inactivate HSCs and reduce the inflammatory response, eventually leading to activation of fibrolysis and degradation of ECM. Thus, it is critical to understand the role of gene-environment interactions in the context of liver fibrosis progression and regression in order to identify specific therapeutic targets for optimized treatment to induce fibrosis regression, prevent HCC development and, ultimately, improve the clinical outcome.
Collapse
Affiliation(s)
- Anindita Banerjee
- Department of Transfusion Transmitted Diseases, ICMR-National Institute of Immunohaematology, Mumbai 400012, Maharashtra, India;
| | - Patrizia Farci
- Hepatic Pathogenesis Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
3
|
Cui G, Liu H, Laugsand JB. Endothelial cells-directed angiogenesis in colorectal cancer: Interleukin as the mediator and pharmacological target. Int Immunopharmacol 2023; 114:109525. [PMID: 36508917 DOI: 10.1016/j.intimp.2022.109525] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/22/2022] [Accepted: 11/27/2022] [Indexed: 12/14/2022]
Abstract
Enhanced angiogenesis is a cancer hallmark and critical for colorectal cancer (CRC) invasion and metastasis. Upon exposure to proangiogenic factors, therefore, targeting tumor-associated proangiogenic factors/receptors hold great promise as a therapeutic modality to treat CRC, particularly metastatic CRC. Accumulating evidence from numerous studies suggests that tumor endothelial cells (ECs) are not only the target of proangiogenic factors, but also function as the cellular source of proangiogenic factors. Studies showed that ECs can produce different proangiogenic factors to participate in the regulation of angiogenesis process, in which ECs-derived interleukins (ILs) show a potential stimulatory effect on angiogenesis via either an direct action on their receptors expressed on progenitor of ECs or an indirect way through enhanced production of other proangiogenic factors. Although a great deal of attention is given to the effects of tumor-derived and immune cell-derived ILs, few studies describe the potential effects of vascular ECs-derived ILs on the tumor angiogenesis process. This review provides an updated summary of available information on proangiogenic ILs, such as IL-1, IL-6, IL-8, IL-17, IL-22, IL-33, IL-34, and IL-37, released by microvascular ECs as potential drivers of the tumor angiogenesis process and discusses their potential as a novel candidate for antiangiogenic target for the treatment of CRC patients.
Collapse
Affiliation(s)
- Guanglin Cui
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Faculty of Health Science, Nord University, Campus Levanger, Norway.
| | - Hanzhe Liu
- School of Stomatology, Wuhan University, Wuhan, China.
| | | |
Collapse
|
4
|
Nisimura LM, Ferreira RR, Coelho LL, de Souza EM, Gonzaga BM, Ferrão PM, Waghabi MC, de Mesquita LB, Pereira MCDS, Moreira ODC, Lannes-Vieira J, Garzoni LR. Increased angiogenesis parallels cardiac tissue remodelling in experimental acute Trypanosoma cruzi infection. Mem Inst Oswaldo Cruz 2022; 117:e220005. [PMID: 36417626 PMCID: PMC9677593 DOI: 10.1590/0074-02760220005] [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: 01/07/2022] [Accepted: 09/16/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Angiogenesis has been implicated in tissue injury in several noninfectious diseases, but its role in Chagas disease (CD) physiopathology is unclear. OBJECTIVES The present study aimed to investigate the effect of Trypanosoma cruzi infection on cardiac angiogenesis during the acute phase of experimental CD. METHODS The signalling pathway involved in blood vessel formation and cardiac remodelling was evaluated in Swiss Webster mice infected with the Y strain of T. cruzi. The levels of molecules involved in the regulation of angiogenesis, such as vascular endothelial growth factor-A (VEGF-A), Flk-1, phosphorylated extracellular-signal-regulated protein kinase (pERK), hypoxia-inducible factor-1α (HIF-1α), CD31, α-smooth muscle actin (α-SMA) and also the blood vessel growth were analysed during T. cruzi infection. Hearts were analysed using conventional histopathology, immunohistochemistry and western blotting. FINDINGS In this study, our data demonstrate that T. cruzi acute infection in mice induces exacerbated angiogenesis in the heart and parallels cardiac remodelling. In comparison with noninfected controls, the cardiac tissue of T. cruzi-infected mice presented higher levels of (i) HIF-1α, VEGF-A, Flk-1 and pERK; (ii) angiogenesis; (iii) α-SMA+ cells in the tissue; and (iv) collagen -1 deposition around blood vessels and infiltrating throughout the myocardium. MAIN CONCLUSIONS We observed cardiac angiogenesis during acute experimental T. cruzi infection parallels cardiac inflammation and remodelling.
Collapse
Affiliation(s)
- Lindice Mitie Nisimura
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, RJ, Brasil
| | - Roberto Rodrigues Ferreira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, RJ, Brasil,Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Genômica Funcional e Bioinformática, Rio de Janeiro, RJ, Brasil
| | - Laura Lacerda Coelho
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, RJ, Brasil
| | - Elen Mello de Souza
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Virologia Molecular, Rio de Janeiro, RJ, Brasil
| | - Beatriz Matheus Gonzaga
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, RJ, Brasil
| | - Patrícia Mello Ferrão
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, RJ, Brasil,Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Genômica Funcional e Bioinformática, Rio de Janeiro, RJ, Brasil
| | - Mariana Caldas Waghabi
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Virologia Molecular, Rio de Janeiro, RJ, Brasil
| | - Liliane Batista de Mesquita
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Ultraestrutura Celular, Rio de Janeiro, RJ, Brasil
| | | | - Otacilio da Cruz Moreira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Biologia Molecular e Doenças Endêmicas, Rio de Janeiro, RJ, Brasil
| | - Joseli Lannes-Vieira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Biologia das Interações, Rio de Janeiro, RJ, Brasil
| | - Luciana Ribeiro Garzoni
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, RJ, Brasil,+ Corresponding author:
| |
Collapse
|
5
|
Monavarian M, Elhaw AT, Tang PW, Javed Z, Shonibare Z, Scalise CB, Arend R, Jolly MK, Sewell-Loftin MK, Hempel N, Mythreye K. Emerging perspectives on growth factor metabolic relationships in the ovarian cancer ascites environment. Semin Cancer Biol 2022; 86:709-719. [PMID: 35259492 PMCID: PMC9441472 DOI: 10.1016/j.semcancer.2022.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 02/07/2023]
Abstract
The ascites ecosystem in ovarian cancer is inhabited by complex cell types and is bathed in an environment rich in cytokines, chemokines, and growth factors that directly and indirectly impact metabolism of cancer cells and tumor associated cells. This milieu of malignant ascites, provides a 'rich' environment for the disease to thrive, contributing to every aspect of advanced ovarian cancer, a devastating gynecological cancer with a significant gap in targeted therapeutics. In this perspective we focus our discussions on the 'acellular' constituents of this liquid malignant tumor microenvironment, and how they influence metabolic pathways. Growth factors, chemokines and cytokines are known modulators of metabolism and have been shown to impact nutrient uptake and metabolic flexibility of tumors, yet few studies have explored how their enrichment in malignant ascites of ovarian cancer patients contributes to the metabolic requirements of ascites-resident cells. We focus here on TGF-βs, VEGF and ILs, which are frequently elevated in ovarian cancer ascites and have all been described to have direct or indirect effects on metabolism, often through gene regulation of metabolic enzymes. We summarize what is known, describe gaps in knowledge, and provide examples from other tumor types to infer potential unexplored roles and mechanisms for ovarian cancer. The distribution and variation in acellular ascites components between patients poses both a challenge and opportunity to further understand how the ascites may contribute to disease heterogeneity. The review also highlights opportunities for studies on ascites-derived factors in regulating the ascites metabolic environment that could act as a unique signature in aiding clinical decisions in the future.
Collapse
Affiliation(s)
- Mehri Monavarian
- Division of Molecular Cellular Pathology, Department of Pathology, O'Neal Comprehensive Cancer Center, University of Alabama Heersink School of Medicine, Birmingham, AL, USA
| | - Amal Taher Elhaw
- Division of Hematology Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh PA 15213, USA
| | - Priscilla W Tang
- Division of Hematology Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh PA 15213, USA
| | - Zaineb Javed
- Division of Hematology Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh PA 15213, USA
| | - Zainab Shonibare
- Division of Molecular Cellular Pathology, Department of Pathology, O'Neal Comprehensive Cancer Center, University of Alabama Heersink School of Medicine, Birmingham, AL, USA
| | - Carly Bess Scalise
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Alabama School of Medicine, Birmingham, AL, USA
| | - Rebecca Arend
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Alabama School of Medicine, Birmingham, AL, USA
| | - Mohit Kumar Jolly
- Center for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, O'Neal Comprehensive Cancer Center, University of Alabama School of Medicine, Birmingham, AL, USA
| | - Nadine Hempel
- Division of Hematology Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh PA 15213, USA.
| | - Karthikeyan Mythreye
- Division of Molecular Cellular Pathology, Department of Pathology, O'Neal Comprehensive Cancer Center, University of Alabama Heersink School of Medicine, Birmingham, AL, USA.
| |
Collapse
|
6
|
Morris RM, Mortimer TO, O’Neill KL. Cytokines: Can Cancer Get the Message? Cancers (Basel) 2022; 14:cancers14092178. [PMID: 35565306 PMCID: PMC9103018 DOI: 10.3390/cancers14092178] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cytokines are important molecular players in cancer development, progression, and potential targets for treatment. Despite being small and overlooked, research has revealed that cytokines influence cancer biology in multiple ways. Cytokines are often found to contribute to immune function, cell damage, inflammation, angiogenesis, metastasis, and several other cellular processes important to tumor survival. Cytokines have also proven to have powerful effects on complex tumor microenvironment molecular biology and microbiology. Due to their heavy involvement in critical cancer-related processes, cytokines have also become attractive therapeutic targets for cancer treatment. In this review, we describe the relationship between several cytokines and crucial cancer-promoting processes and their therapeutic potential. Abstract Cytokines are small molecular messengers that have profound effects on cancer development. Increasing evidence shows that cytokines are heavily involved in regulating both pro- and antitumor activities, such as immune activation and suppression, inflammation, cell damage, angiogenesis, cancer stem-cell-like cell maintenance, invasion, and metastasis. Cytokines are often required to drive these cancer-related processes and, therefore, represent an important research area for understanding cancer development and the potential identification of novel therapeutic targets. Interestingly, some cytokines are reported to be related to both pro- and anti-tumorigenicity, indicating that cytokines may play several complex roles relating to cancer pathogenesis. In this review, we discuss some major cancer-related processes and their relationship with several cytokines.
Collapse
|
7
|
Liu XQ, Shao XR, Liu Y, Dong ZX, Chan SH, Shi YY, Chen SN, Qi L, Zhong L, Yu Y, Lv T, Yang PF, Li LY, Wang XB, Zhang XD, Li X, Zhao W, Sehgal L, Li M, Zhang XD. Tight junction protein 1 promotes vasculature remodeling via regulating USP2/TWIST1 in bladder cancer. Oncogene 2022; 41:502-514. [PMID: 34782718 DOI: 10.1038/s41388-021-02112-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 10/27/2021] [Accepted: 11/01/2021] [Indexed: 12/15/2022]
Abstract
Bladder cancer (BLCA) is the most common malignant tumor of the urinary system and is characterized by high metastatic rates and poor prognosis. The expression of tight junction protein 1 (TJP1) is associated with bladder cancer invasion; however, the mechanism by which TJP1 affects vasculature remodeling remains unknown. In this study, we found that TJP1 expression correlated with tumor angiogenesis and poor overall survival in clinical samples. Furthermore, TJP1 overexpression promoted tumor angiogenesis in BLCA cells and stimulated recruitment of macrophages to tumors by upregulating CCL2 expression. Mechanistically, TJP1 interacted with TWIST1 and enhanced the transcriptional activity of CCL2. The impairment of tumor angiogenesis caused by knockdown of TJP1 was dramatically rescued by overexpression of TWIST1. Furthermore, TJP1 recruited USP2, which deubiquitinated TWIST1, thereby protecting TWIST1 from proteasome-mediated protein degradation. In conclusion, our results suggest that TJP1 controls angiogenesis in BLCA via TWIST1-dependent regulation of CCL2. We demonstrate that TJP1 functions as a scaffold for the interaction between USP2 and TWIST1 and this may provide potential therapeutic targets in bladder cancer.
Collapse
Affiliation(s)
- Xue-Qi Liu
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xin-Rong Shao
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Ye Liu
- Department of Pathology, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Zhao-Xia Dong
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Sze-Hoi Chan
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Yuan-Yuan Shi
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Shu-Na Chen
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Lin Qi
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Li Zhong
- State Key Laboratory of Oncology in South China, Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yue Yu
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Ting Lv
- Department of Pathology, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Peng-Fei Yang
- Department of Pathology, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Li-Yan Li
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xiao-Bin Wang
- Andrology section, Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xu-Dong Zhang
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xin Li
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Wenxue Zhao
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Lalit Sehgal
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Miao Li
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China. .,Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, China.
| | - Xing-Ding Zhang
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
8
|
Kim H, Shin Y, Kim DH. Mechanobiological Implications of Cancer Progression in Space. Front Cell Dev Biol 2021; 9:740009. [PMID: 34957091 PMCID: PMC8692837 DOI: 10.3389/fcell.2021.740009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/18/2021] [Indexed: 12/11/2022] Open
Abstract
The human body is normally adapted to maintain homeostasis in a terrestrial environment. The novel conditions of a space environment introduce challenges that changes the cellular response to its surroundings. Such an alteration causes physical changes in the extracellular microenvironment, inducing the secretion of cytokines such as interleukin-6 (IL-6) and tumor growth factor-β (TGF-β) from cancer cells to enhance cancer malignancy. Cancer is one of the most prominent cell types to be affected by mechanical cues via active interaction with the tumor microenvironment. However, the mechanism by which cancer cells mechanotransduce in the space environment, as well as the influence of this process on human health, have not been fully elucidated. Due to the growing interest in space biology, this article reviews cancer cell responses to the representative conditions altered in space: microgravity, decompression, and irradiation. Interestingly, cytokine and gene expression that assist in tumor survival, invasive phenotypic transformation, and cancer cell proliferation are upregulated when exposed to both simulated and actual space conditions. The necessity of further research on space mechanobiology such as simulating more complex in vivo experiments or finding other mechanical cues that may be encountered during spaceflight are emphasized.
Collapse
Affiliation(s)
- Hyondeog Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea
| | - Yun Shin
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea.,Department of Integrative Energy Engineering, College of Engineering, Korea University, Seoul, South Korea
| |
Collapse
|
9
|
Toboni MD, Lomonosova E, Bruce SF, Tankou JI, Mullen MM, Schab A, Oplt A, Noia H, Wilke D, Kuroki LM, Hagemann AR, McCourt CK, Thaker PH, Powell MA, Khabele D, Mutch DG, Fuh KC. Inhibition of AXL and VEGF-A Has Improved Therapeutic Efficacy in Uterine Serous Cancer. Cancers (Basel) 2021; 13:5877. [PMID: 34884986 PMCID: PMC8656641 DOI: 10.3390/cancers13235877] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/05/2021] [Accepted: 11/16/2021] [Indexed: 11/17/2022] Open
Abstract
Endometrial cancer remains the most prevalent gynecologic cancer with continued rising incidence. A less common form of this cancer is uterine serous cancer, which represents 10% of endometrial cancer cases. However, this is the most aggressive cancer. The objective was to assess whether inhibiting the receptor tyrosine kinase AXL with AVB-500 in combination with bevacizumab would improve response in uterine serous cancer. To prove this, we conducted multiple angiogenesis assays including tube formation assays and angiogenesis invasion assays. In addition, we utilized mouse models with multiple cells lines and subsequently analyzed harvested tissue through immunohistochemistry CD31 staining to assess microvessel density. The combination treatment arms demonstrated decreased angiogenic potential in each assay. In addition, intraperitoneal mouse models demonstrated a significant decrease in tumor burden in two cell lines. The combination of AVB-500 and bevacizumab reduced tumor burden in vivo and reduced morphogenesis and migration in vitro which are vital to the process of angiogenesis.
Collapse
Affiliation(s)
- Michael D. Toboni
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
| | - Elena Lomonosova
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Shaina F. Bruce
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
| | - Jo’an I. Tankou
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
| | - Mary M. Mullen
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
| | - Angela Schab
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Alyssa Oplt
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Hollie Noia
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Danny Wilke
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Lindsay M. Kuroki
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
| | - Andrea R. Hagemann
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
| | - Carolyn K. McCourt
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
| | - Premal H. Thaker
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
| | - Matthew A. Powell
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
| | - Dineo Khabele
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
| | - David G. Mutch
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
| | - Katherine C. Fuh
- Barnes Jewish Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, MO 63110, USA; (M.D.T.); (E.L.); (S.F.B.); (J.I.T.); (M.M.M.); (A.S.); (A.O.); (H.N.); (D.W.); (L.M.K.); (A.R.H.); (C.K.M.); (P.H.T.); (M.A.P.); (D.K.); (D.G.M.)
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| |
Collapse
|
10
|
Sakamoto N, Kurokawa R, Watadani T, Morikawa T, Nakaya M, Cho S, Fujita N, Kamio S, Koyama H, Suzuki S, Yamada H, Abe O, Gonoi W. Differential diagnosis of thymic epithelial neoplasms on computed tomography using the diameter of the thymic vein. Medicine (Baltimore) 2021; 100:e27942. [PMID: 34797351 PMCID: PMC8601265 DOI: 10.1097/md.0000000000027942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 01/05/2023] Open
Abstract
Although differentiating benign and malignant thymic epithelial lesions is important to avoid unnecessary treatment and predict prognosis, it is challenging because of overlaps in the chest computed tomography (CT) findings. In this study, we investigated whether the diameter of the thymic vein and other CT findings could differentiate between benign (thymoma and thymic cysts) and malignant (thymic carcinoma, [TCa]) lesions.We conducted a retrospective study across two tertiary referral hospitals in Japan between November 2009 and June 2018. We included 12 patients with TCa, 34 patients with thymomas, and 17 patients with thymic cysts. We analyzed the receiver operating characteristic (ROC) curve to determine the best cut-off values and performed univariate and multivariate analyses of CT findings to distinguish TCa from other benign lesions. Post-hoc analysis was performed for the maximum short axis of the thymic vein using the Mann-Whitney U test, and the number of the maximum short axis of the thymic vein ≥ the cutoff was determined using the Fisher exact test with a family-wise error-correction using Bonferroni's method.ROC analysis showed that a maximum short axis of the thymic vein ≥2 mm was considerably more frequent in TCa than in the other lesions (P < .001 for both), with 83% sensitivity and 86% specificity. Univariate and multivariate analyses revealed the association with TCa of the number of the maximum short axis of the thymic vein ≥2 mm (P = .005, multivariate generalized linear model analysis), ill-defined margin (P = .001), and mediastinal lymphadenopathy (P < .001). Thymic vein diameter was in descendimg order of TCa > thymoma > thymic cysts with statistically significant differences between the groups (Ps < .05).Thymic vein diameter was significantly longer in TCa than in thymoma and thymic cysts. Measurement of the maximum short axis of the thymic vein could be a powerful diagnostic tool to differentiate TCa from thymoma and thymic cysts.
Collapse
Affiliation(s)
- Naoya Sakamoto
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Ryo Kurokawa
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Takeyuki Watadani
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Teppei Morikawa
- Department of Diagnostic Pathology, NTT Medical Center Tokyo, 5-9-22 Higashi-Gotanda, Shinagawa-ku, Tokyo, Japan
| | - Moto Nakaya
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Shinichi Cho
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Nana Fujita
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Satoru Kamio
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Hiroaki Koyama
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Satoshi Suzuki
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Haruyasu Yamada
- Department of Radiology, NTT Medical Center Tokyo, 5-9-22 Higashi-Gotanda, Shinagawa-ku, Tokyo, Japan
| | - Osamu Abe
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Wataru Gonoi
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
11
|
Stoycheva D, Simsek H, Weber W, Hauser AE, Klotzsch E. External cues to drive B cell function towards immunotherapy. Acta Biomater 2021; 133:222-230. [PMID: 33636402 DOI: 10.1016/j.actbio.2021.02.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/31/2022]
Abstract
Immunotherapy stands out as a powerful and promising therapeutic strategy in the treatment of cancer, infections, and autoimmune diseases. Adoptive immune therapies are usually centered on modified T cells and their specific expansion towards antigen-specific T cells against cancer and other diseases. However, despite their unmatched features, the potential of B cells in immunotherapy is just beginning to be explored. The main role of B cells in the immune response is to secrete antigen-specific antibodies and provide long-term protection against foreign pathogens. They further function as antigen-presenting cells (APCs) and secrete pro- and anti-inflammatory cytokines and thus exert positive and negative regulatory stimuli on other cells involved in the immune response such as T cells. Therefore, while hyperactivation of B cells can cause autoimmunity, their dysfunctions lead to severe immunodeficiencies. Only suitably activated B cells can play an active role in the treatment of cancers, infections, and autoimmune diseases. As a result, studies have focused on B cell-targeted immunotherapies in recent years. For this, the development, functions, interactions with the microenvironment, and clinical importance of B cells should be well understood. In this review, we summarize the main events during B cell activation. From the viewpoint of mechanobiology we discuss the translation of external cues such as surface topology, substrate stiffness, and biochemical signaling into B cell functions. We further dive into current B cell-targeted therapy strategies and their clinical applications. STATEMENT OF SIGNIFICANCE: B cells are proving as a promising tool in the field of immunotherapy. B cells exhibit various functions such as antibody production, antigen presentation or secretion of immune-regulatory factors which can be utilized in the fight against oncological or immunological disorders. In this review we discuss the importance of external mechanobiological cues such as surface topology, substrate stiffness, and biochemical signaling on B cell function. We further summarize B cell-targeted therapy strategies and their clinical applications, as in the context of anti-tumor responses and autoimmune diseases.
Collapse
|
12
|
Sarogni P, Mapanao AK, Marchetti S, Kusmic C, Voliani V. A Standard Protocol for the Production and Bioevaluation of Ethical In Vivo Models of HPV-Negative Head and Neck Squamous Cell Carcinoma. ACS Pharmacol Transl Sci 2021; 4:1227-1234. [PMID: 34151212 PMCID: PMC8205242 DOI: 10.1021/acsptsci.1c00083] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Indexed: 11/29/2022]
Abstract
Preclinical cancer research increasingly demands sophisticated models for the development and translation of efficient and safe cancer treatments to clinical practice. In this regard, tumor-grafted chorioallantoic membrane (CAM) models are biological platforms that account for the dynamic roles of the tumor microenvironment and cancer physiopathology, allowing straightforward investigations in agreement to the 3Rs concept (the concept of reduction, refinement, and replacement of animal models). CAM models are the next advanced model for tumor biological explorations as well as for reliable assessment regarding initial efficacy, toxicity, and systemic biokinetics of conventional and emerging neoplasm treatment modalities. Here we report a standardized and optimized protocol for the production and biocharacterization of human papillomavirus (HPV)-negative head and neck chick chorioallantoic membrane models from a commercial cell line (SCC-25). Oral malignancies continue to have severe morbidity with less than 50% long-term survival despite the advancement in the available therapies. Thus, there is a persisting demand for new management approaches to establish more efficient strategies toward their treatment. Remarkably, the inclusion of CAM models in the preclinical research workflow is crucial to ethically foster both the basic and translational oncological research on oral malignancies as well as for the advancement of efficient cancer treatment approaches.
Collapse
Affiliation(s)
- Patrizia Sarogni
- Center
for Nanotechnology Innovation@NEST, Istituto
Italiano di Tecnologia, Piazza San Silvestro 12, Pisa 56126, Italy
| | - Ana Katrina Mapanao
- Center
for Nanotechnology Innovation@NEST, Istituto
Italiano di Tecnologia, Piazza San Silvestro 12, Pisa 56126, Italy
- NEST-Scuola
Normale Superiore, Piazza
San Silvestro 12, Pisa 56126, Italy
| | - Sabrina Marchetti
- Institute
of Clinical Physiology, CNR, Via G. Moruzzi 1, Pisa 56100, Italy
| | - Claudia Kusmic
- Institute
of Clinical Physiology, CNR, Via G. Moruzzi 1, Pisa 56100, Italy
| | - Valerio Voliani
- Center
for Nanotechnology Innovation@NEST, Istituto
Italiano di Tecnologia, Piazza San Silvestro 12, Pisa 56126, Italy
| |
Collapse
|
13
|
Kumar R, Harilal S, Parambi DGT, Narayanan SE, Uddin MS, Marathakam A, Jose J, Mathew GE, Mathew B. Fascinating Chemopreventive Story of Wogonin: A Chance to Hit on the Head in Cancer Treatment. Curr Pharm Des 2021; 27:467-478. [PMID: 32338206 DOI: 10.2174/1385272824999200427083040] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/30/2020] [Indexed: 11/22/2022]
Abstract
Cancer, global havoc, is a group of debilitating diseases that strikes family as well as society. Cancer cases are drastically increasing these days. Despite many therapies and surgical procedures available, cancer is still difficult to control due to limited effective therapies or targeted therapies. Natural products can produce lesser side effects to the normal cells, which are the major demerit of chemotherapies and radiation. Wogonin, a natural product extracted from the plant, Scutellaria baicalensis has been widely studied and found with a high caliber to tackle most of the cancers via several mechanisms that include intrinsic as well as extrinsic apoptosis signaling pathways, carcinogenesis diminution, telomerase activity inhibition, metastasis inhibition in the inflammatory microenvironment, anti-angiogenesis, cell growth inhibition and arrest of the cell cycle, increased generation of H2O2 and accumulation of Ca2+ and also as an adjuvant along with anticancer drugs. This article discusses the role of wogonin in various cancers, its synergism with various drugs, and the mechanism by which wogonin controls tumor growth.
Collapse
Affiliation(s)
- Rajesh Kumar
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Seetha Harilal
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Della G T Parambi
- College of Pharmacy, Department of Pharmaceutical Chemistry, Jouf University, Sakaka, Al Jouf, 2014, Saudi Arabia
| | - Siju E Narayanan
- P.G. Department of Pharmacology, College of Pharmaceutical Sciences, Government Medical College, Kannur-670503, India
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Akash Marathakam
- Department of Pharmaceutical Chemistry, National College of Pharmacy, Calicut, India
| | - Jobin Jose
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Science, NITTE Deemed to be University, Manglore, 575018, India
| | - Githa E Mathew
- Department of Pharmacology, Grace College of Pharmacy, Palakkad, India
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, 678557, Kerala, India
| |
Collapse
|
14
|
Abstract
Since the initial reports implicating caveolin-1 (CAV1) in neoplasia, the scientific community has made tremendous strides towards understanding how CAV1-dependent signaling and caveolae assembly modulate solid tumor growth. Once a solid neoplastic tumor reaches a certain size, it will increasingly rely on its stroma to meet the metabolic demands of the rapidly proliferating cancer cells, a limitation typically but not exclusively addressed via the formation of new blood vessels. Landmark studies using xenograft tumor models have highlighted the importance of stromal CAV1 during neoplastic blood vessel growth from preexisting vasculature, a process called angiogenesis, and helped identify endothelium-specific signaling events regulated by CAV1, such as vascular endothelial growth factor (VEGF) receptors as well as the endothelial nitric oxide (NO) synthase (eNOS) systems. This chapter provides a glimpse into the signaling events modulated by CAV1 and its scaffolding domain (CSD) during endothelial-specific aspects of neoplastic growth, such as vascular permeability, angiogenesis, and mechanotransduction.
Collapse
Affiliation(s)
- Pascal Bernatchez
- Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, University of British Columbia (UBC), 2176 Health Sciences mall, room 217, Vancouver, BC, V6T 1Z3, Canada. .,Centre for Heart & Lung Innovation, St. Paul's Hospital, Vancouver, Canada.
| |
Collapse
|
15
|
Extract from the Coriolus versicolor Fungus as an Anti-Inflammatory Agent with Cytotoxic Properties against Endothelial Cells and Breast Cancer Cells. Int J Mol Sci 2020; 21:ijms21239063. [PMID: 33260615 PMCID: PMC7731170 DOI: 10.3390/ijms21239063] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic inflammation is a well-recognised tumour-enabling component, which includes bioactive molecules from cells infiltrating the tumour microenvironment and increases the risk of cancer progression. Since long-term use of the currently available anti-inflammatory drugs used in cancer therapy causes numerous side effects, the aim of this study was to investigate the effect of an extract isolated from the Coriolus versicolor fungus (CV extract) on HUVEC endothelial cells and MCF-7 breast cancer cells in a pro-inflammatory microenvironment mimicked by lipopolysaccharide (LPS). The cells were simultaneously stimulated with the LPS and CV extract. After co-treatment, the cell viability, generation of reactive oxygen species (ROS), wound-healing assay, production of the pro-inflammatory and pro-angiogenic factors (interleukin (IL) 6, IL-8, and metalloproteinase (MMP) 9)), as well as expression of Toll-like receptor (TLR) 4 and phosphorylated IκB (p-IκB) were evaluated. The results showed that the CV extract inhibited IL-6, IL-8, and MMP-9 production by the LPS-stimulated cells. This effect was accompanied by a decrease in TLR4 and p-IκB expression. The CV extract also had anti-migratory properties and induced a cytotoxic effect on the cells that was enhanced in the presence of LPS. The observed cytotoxicity was associated with an increase in ROS generation. We conclude that the CV extract possesses cytotoxic activity against cancer cells and endothelial cells and has the ability to inhibit the expression of the pro-tumorigenic factors associated with inflammation.
Collapse
|
16
|
Virumbrales-Muñoz M, Chen J, Ayuso J, Lee M, Abel EJ, Beebe DJ. Organotypic primary blood vessel models of clear cell renal cell carcinoma for single-patient clinical trials. LAB ON A CHIP 2020; 20:4420-4432. [PMID: 33103699 PMCID: PMC8743028 DOI: 10.1039/d0lc00252f] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common genitourinary cancer associated with the development of abnormal tumor angiogenesis. Although multiple anti-angiogenic therapies have been developed, responses to individual treatment are highly variable between patients. Thus, the use of one-patient clinical trials has been suggested as an alternative to standard trials. We used a microfluidic device to generate organotypic primary patient-specific blood vessel models using normal (NEnC) and tumor-associated primary CD31+ selected cells (TEnC). Our model was able to recapitulate differences in angiogenic sprouting and vessel permeability that characterize normal and tumor-associated vessels. We analyzed the expression profile of vessel models to define vascular normalization in a patient-specific manner. Using this data, we identified actionable targets to normalize TEnC vessel function to a more NEnC-like phenotype. Finally, we tested two of these drugs in our patient-specific models to determine the efficiency in restoring vessel function showing the potential of the model for single-patient clinical trials.
Collapse
Affiliation(s)
- María Virumbrales-Muñoz
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, USA.
| | | | | | | | | | | |
Collapse
|
17
|
Abstract
Microvasculature functions at the tissue and cell level, regulating local mass exchange of oxygen and nutrient-rich blood. While there has been considerable success in the biofabrication of large- and small-vessel replacements, functional microvasculature has been particularly challenging to engineer due to its size and complexity. Recently, three-dimensional bioprinting has expanded the possibilities of fabricating sophisticated microvascular systems by enabling precise spatiotemporal placement of cells and biomaterials based on computer-aided design. However, there are still significant challenges facing the development of printable biomaterials that promote robust formation and controlled 3D organization of microvascular networks. This review provides a thorough examination and critical evaluation of contemporary biomaterials and their specific roles in bioprinting microvasculature. We first provide an overview of bioprinting methods and techniques that enable the fabrication of microvessels. We then offer an in-depth critical analysis on the use of hydrogel bioinks for printing microvascularized constructs within the framework of current bioprinting modalities. We end with a review of recent applications of bioprinted microvasculature for disease modeling, drug testing, and tissue engineering, and conclude with an outlook on the challenges facing the evolution of biomaterials design for bioprinting microvasculature with physiological complexity.
Collapse
Affiliation(s)
- Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sophia E. Silver
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael Yost
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
18
|
van de Veen W, Globinska A, Jansen K, Straumann A, Kubo T, Verschoor D, Wirz OF, Castro-Giner F, Tan G, Rückert B, Ochsner U, Herrmann M, Stanić B, van Splunter M, Huntjens D, Wallimann A, Fonseca Guevara RJ, Spits H, Ignatova D, Chang YT, Fassnacht C, Guenova E, Flatz L, Akdis CA, Akdis M. A novel proangiogenic B cell subset is increased in cancer and chronic inflammation. SCIENCE ADVANCES 2020; 6:eaaz3559. [PMID: 32426497 PMCID: PMC7220305 DOI: 10.1126/sciadv.aaz3559] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 03/06/2020] [Indexed: 05/17/2023]
Abstract
B cells contribute to immune responses through the production of immunoglobulins, antigen presentation, and cytokine production. Several B cell subsets with distinct functions and polarized cytokine profiles have been reported. In this study, we used transcriptomics analysis of immortalized B cell clones to identify an IgG4+ B cell subset with a unique function. These B cells are characterized by simultaneous expression of proangiogenic cytokines including VEGF, CYR61, ADM, FGF2, PDGFA, and MDK. Consequently, supernatants from these clones efficiently promote endothelial cell tube formation. We identified CD49b and CD73 as surface markers identifying proangiogenic B cells. Circulating CD49b+CD73+ B cells showed significantly increased frequency in patients with melanoma and eosinophilic esophagitis (EoE), two diseases associated with angiogenesis. In addition, tissue-infiltrating IgG4+CD49b+CD73+ B cells expressing proangiogenic cytokines were detected in patients with EoE and melanoma. Our results demonstrate a previously unidentified proangiogenic B cell subset characterized by expression of CD49b, CD73, and proangiogenic cytokines.
Collapse
Affiliation(s)
- Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Anna Globinska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Kirstin Jansen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Alex Straumann
- Swiss EoE Clinic and EoE Research Network, Olten, Switzerland
| | - Terufumi Kubo
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Daniëlle Verschoor
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Oliver F. Wirz
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Francesc Castro-Giner
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Functional Genomics Center Zurich, ETH Zurich/University of Zurich, Zurich, Switzerland
| | - Ge Tan
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Functional Genomics Center Zurich, ETH Zurich/University of Zurich, Zurich, Switzerland
| | - Beate Rückert
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Urs Ochsner
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Marietta Herrmann
- AO Research Institute Davos, Davos, Switzerland
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Barbara Stanić
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Marloes van Splunter
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Daan Huntjens
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Alexandra Wallimann
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- AO Research Institute Davos, Davos, Switzerland
| | | | - Hergen Spits
- AIMM Therapeutics, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam Medical Centers, Amsterdam, Netherlands
| | - Desislava Ignatova
- Department of Dermatology, University Hospital Zurich, University of Zurich, Switzerland
| | - Yun-Tsan Chang
- Department of Dermatology, University Hospital Zurich, University of Zurich, Switzerland
| | - Christina Fassnacht
- Department of Dermatology, University Hospital Zurich, University of Zurich, Switzerland
| | - Emmanuella Guenova
- Department of Dermatology, University Hospital Zurich, University of Zurich, Switzerland
- University Hospital of Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St. Gallen, Switzerland
- Department of Oncology and Haematology, Kantonsspital St. Gallen, Switzerland
- Department of Dermatology and Allergology, Kantonsspital St. Gallen, Switzerland
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Corresponding author.
| |
Collapse
|
19
|
Ciciola P, Cascetta P, Bianco C, Formisano L, Bianco R. Combining Immune Checkpoint Inhibitors with Anti-Angiogenic Agents. J Clin Med 2020; 9:E675. [PMID: 32138216 PMCID: PMC7141336 DOI: 10.3390/jcm9030675] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy has recently emerged as a novel strategy for treating different types of solid tumors, with promising results. However, still a large fraction of patients do not primarily respond to such approaches, and even responders sooner or later develop resistance. Moreover, immunotherapy is a promising strategy for certain malignancies but not for others, with this discrepancy having been attributed to a more immunogenic microenvironment of some tumors. As abnormal and augmented tumor vessels often occur in cancerogenesis, anti-angiogenic drugs have already demonstrated their effectiveness both in preclinical and in clinical settings. By targeting abnormal formation of tumor vessels, anti-angiogenetic agents potentially result in an enhanced infiltration of immune effector cells. Moreover, crosstalks downstream of the immune checkpoint axis and vascular endothelial growth factor receptor (VEGFR) signaling may result in synergistic effects of combined treatment in tumor cells. In this review, we will describe and discuss the biological rationale of a combined therapy, underlying the modification in tumor microenvironment as well as in tumor cells after exposure to checkpoint inhibitors and anti-angiogenic drugs. Moreover, we will highlight this strategy as a possible way for overcoming drug resistance. By first discussing potential prognostic and predictive factors for combined treatment, we will then turn to clinical settings, focusing on clinical trials where this strategy is currently being investigated.
Collapse
Affiliation(s)
- Paola Ciciola
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (P.C.); (P.C.)
| | - Priscilla Cascetta
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (P.C.); (P.C.)
| | - Cataldo Bianco
- Department of Experimental and Clinical Medicine, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy;
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (P.C.); (P.C.)
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (P.C.); (P.C.)
| |
Collapse
|
20
|
Van Cutsem E, Muro K, Cunningham D, Bodoky G, Sobrero A, Cascinu S, Ajani J, Oh SC, Al-Batran SE, Wainberg ZA, Wijayawardana SR, Melemed S, Ferry D, Hozak RR, Ohtsu A. Biomarker analyses of second-line ramucirumab in patients with advanced gastric cancer from RAINBOW, a global, randomized, double-blind, phase 3 study. Eur J Cancer 2020; 127:150-157. [PMID: 32014812 DOI: 10.1016/j.ejca.2019.10.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/21/2019] [Accepted: 10/27/2019] [Indexed: 01/11/2023]
Abstract
BACKGROUND The RAINBOW trial showed that second-line ramucirumab with paclitaxel prolongs overall survival (OS) and progression-free survival (PFS) compared with placebo plus paclitaxel for treatment of advanced gastric/gastroesophageal junction cancer. Plasma samples were collected from patients during the trial and tested to identify predictive and prognostic biomarkers. PATIENTS AND METHODS Circulating factors in plasma samples from mutually exclusive subsets of RAINBOW patients were assayed using: Intertek assays (24 markers, 380 samples, 57% of patients) and Lilly-developed assay (LDA) platform (5 markers, 257 samples, 39% of patients). Time-trend plots were generated for each marker from the Intertek assays. Baseline patient data were dichotomized into low- and high-marker subgroups. Markers were analyzed for predictive effects using interaction models and for prognostic effects using main-effects models. RESULTS The Intertek and LDA populations were representative of the full trial population. Plasma levels of VEGF-D and PlGF increased from baseline levels during treatment, then declined after treatment discontinued. Angiopoietin-2 exhibited a decrease during treatment, then increased after treatment discontinuation. No clear time trend was evident with the other markers. Analyses of baseline biomarker expression and its relationship with efficacy variables found no biomarker was predictive for efficacy outcomes, including VEGF-D. However, CRP, HGF, ICAM-3, IL-8, SAA, and VCAM-1 were identified as potential prognostic markers with low baseline levels corresponding to longer OS and PFS. CONCLUSIONS Pharmacodynamic and prognostic relationships were found from the exploratory biomarker analyses in RAINBOW; however, no predictive markers for ramucirumab in gastric cancer were identified in this trial.
Collapse
Affiliation(s)
- E Van Cutsem
- Digestive Oncology, University Hospitals Gasthuisberg, Leuven and KULeuven, Leuven, Belgium.
| | - K Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | | | - G Bodoky
- Department of Oncology, St. László Hospital, Budapest, Hungary
| | - A Sobrero
- Medical Oncology, IRCCS Ospedale San Martino IST, Genova, Italy
| | - S Cascinu
- Department of Medical Oncology, Università Politecnica Delle Marche, Ancona, Italy
| | - J Ajani
- Department of Gastrointestinal Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - S C Oh
- Korea University Guro Hospital, Seoul, South Korea
| | - S E Al-Batran
- Institute of Clinical Cancer Research (IKF), UCT- University Cancer Center, Frankfurt, Germany
| | - Z A Wainberg
- Medical Hematology and Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | | | - S Melemed
- Eli Lilly and Company, Indianapolis, IN, USA
| | - D Ferry
- Eli Lilly and Company, Bridgewater, NJ, USA
| | - R R Hozak
- Eli Lilly and Company, Indianapolis, IN, USA
| | - A Ohtsu
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
21
|
Łasińska I, Kolenda T, Teresiak A, Lamperska KM, Galus Ł, Mackiewicz J. Immunotherapy in Patients with Recurrent and Metastatic Squamous Cell Carcinoma of the Head and Neck. Anticancer Agents Med Chem 2019; 19:290-303. [PMID: 30198439 DOI: 10.2174/1871520618666180910092356] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/09/2018] [Accepted: 07/04/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is the most common malignant cancer occurring in the head and neck area, approximately 90% of the cases. Even in the cases of primary radical treatment (surgical, concomitant chemoradiotherapy or radiotherapy alone), subsequent local recurrence or distant metastases are often observed. In patients with recurrent disease who are unable to receive radical treatment, the results of palliative chemotherapy are not satisfactory. In this review, we summarized the standard treatment options, current development of new drugs and future perspectives in the treatment of patients with recurrent locally advanced and/or metastatic HNSCC. METHODS PubMed databases with words 'head and neck cancer treatment', 'immunotherapy in head and neck cancer treatment' were searched and yielded 186512 and 2249 papers respectively. We selected the most cited articles and reports presenting new immunotherapy agents and drug combinations in HNSCC. RESULTS Recently, two new agents been approved in the treatment of recurrent locally advanced and/or metastatic HNSCC. These are immune-checkpoint inhibitors targeting PD1 (nivolumab and pembrolizumab) which are the most active drugs in the second line treatment of advanced HNSCC. Still, the first line 'golden standard' is the chemotherapy regimen (cisplatin, 5-fluorouracyl) combined with cetuximab. Many phase 3 studies are currently ongoing, evaluating the efficacy of combinational treatment-anti-CTLA4 with anti-PD1 or anti-PDL1. Very encouraging results have been shown in early phase studies evaluating the combination of immunecheckpoint inhibitors with tumor microenvironment immunosuppressive inhibitors. CONCLUSION Despite the huge progress in the systemic treatment of patients with recurrent locally advanced and/or metastatic HNSCC, the disease at this stage remains incurable. Undoubtedly, further research in the field of biomarkers for effective immunotherapy is needed in order to select a group of patients whose will benefit from this therapy, as the treatment is still ineffective in most patients.
Collapse
Affiliation(s)
- Izabela Łasińska
- Department of Medical and Experimental Oncology, Heliodor Swiecicki Clinical Hospital, University of Medical Sciences, Poznan, Poland
| | - Tomasz Kolenda
- Laboratory of Cancer Genetics, Greater Poland Cancer Centre, Poznan, Poland.,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland.,Chair of Medical Biotechnology, University of Medical Sciences, Poznan, Poland
| | - Anna Teresiak
- Laboratory of Cancer Genetics, Greater Poland Cancer Centre, Poznan, Poland
| | | | - Łukasz Galus
- Department of Medical and Experimental Oncology, Heliodor Swiecicki Clinical Hospital, University of Medical Sciences, Poznan, Poland.,Department of Chemotherapy, Greater Poland Cancer Centre, Poznan, Poland
| | - Jacek Mackiewicz
- Department of Medical and Experimental Oncology, Heliodor Swiecicki Clinical Hospital, University of Medical Sciences, Poznan, Poland.,Department of Biology and Environmental Studies, University of Medical Sciences, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland
| |
Collapse
|
22
|
Belenahalli Shekarappa S, Kandagalla S, H Malojirao V, G.S PK, B.T P, Hanumanthappa M. A systems biology approach to identify the key targets of curcumin and capsaicin that downregulate pro-inflammatory pathways in human monocytes. Comput Biol Chem 2019; 83:107162. [DOI: 10.1016/j.compbiolchem.2019.107162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/25/2019] [Accepted: 11/07/2019] [Indexed: 12/17/2022]
|
23
|
Biological correlates of tumor perfusion and its heterogeneity in newly diagnosed breast cancer using dynamic first-pass 18F-FDG PET/CT. Eur J Nucl Med Mol Imaging 2019; 47:1103-1115. [DOI: 10.1007/s00259-019-04422-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/01/2019] [Indexed: 12/30/2022]
|
24
|
Zhang Y, Cheng H, Li W, Wu H, Yang Y. Highly-expressed P2X7 receptor promotes growth and metastasis of human HOS/MNNG osteosarcoma cells via PI3K/Akt/GSK3β/β-catenin and mTOR/HIF1α/VEGF signaling. Int J Cancer 2019; 145:1068-1082. [PMID: 30761524 PMCID: PMC6618011 DOI: 10.1002/ijc.32207] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/06/2018] [Accepted: 01/31/2019] [Indexed: 12/29/2022]
Abstract
The P2X7 receptor, an ATP‐gated ion channel, is critical for cancer cell growth, invasiveness, and angiogenesis. Previous studies indicate that P2X7 regulates osteoblast proliferation and osteodeposition and that high P2X7 expression has a pro‐growth effect in osteosarcoma. However, how it functions in osteosarcoma cell growth and metastasis is not clear. Thus, we elucidated molecular mechanisms of P2X7‐dependent positive regulation of osteosarcoma cell proliferation, invasion, migration, epithelial to mesenchymal transition (EMT), and angiogenesis using in vitro and in vivo models. We confirm that P2X7 is highly‐expressed in human osteosarcoma tumor tissues and HOS/MNNG, MG63, U2OS, SW1353 and SAOS‐2 cell lines. P2X7 receptor stimulation enhanced HOS/MNNG and SAOS‐2 cell proliferation, migration and invasion; but knockdown of P2X7 expression or receptor inhibition had opposite effects. P2X7 positively regulated glycogen content, epithelial to mesenchymal transition and stemness of HOS/MNNG cells. P2X7 activation promoted PI3K/Akt/GSK3β/β‐catenin and mTOR/HIF1α/VEGF signaling, thereby mediating pro‐tumor effects of osteosarcoma cells. Consistent with data from in vitro experiments, systemic administration of P2X7 agonist induced tumor growth, metastasis and tumor‐associated bone destruction in osteosarcoma‐bearing nude mice, whereas a P2X7 antagonist reversed these effects. Thus, the P2X7 receptor participates in regulation of osteosarcoma growth and metastasis and we offer evidence that P2X7 may be a promising therapeutic target for treating osteosarcoma. What's new? The ATP‐gated ion channel receptor P2X7 is increasingly recognized as a tumor‐promoting factor. In this study, P2X7 was found to be overexpressed in human osteosarcoma tissues and cells, with its activation enhancing osteosarcoma cell proliferation, migration, and invasion. P2X7 activation further induced epithelial‐mesenchymal transition (EMT), affected the stemness of osteosarcoma cells, and augmented angiogenesis. Experiments in mice showed that P2X7 also induces osteosarcoma‐associated bone destruction. Opposing effects were observed upon P2X7 inhibition. P2X7 activity was influenced in part by PI3K/Akt/GSK3β/β‐catenin and mTOR/HIF1α/VEGF signaling pathways. The data identify P2X7 as a target for the development of novel therapeutics against osteosarcoma.
Collapse
Affiliation(s)
- Yingchi Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenkai Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Friedman JR, Richbart SD, Merritt JC, Brown KC, Nolan NA, Akers AT, Lau JK, Robateau ZR, Miles SL, Dasgupta P. Acetylcholine signaling system in progression of lung cancers. Pharmacol Ther 2019; 194:222-254. [PMID: 30291908 PMCID: PMC6348061 DOI: 10.1016/j.pharmthera.2018.10.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The neurotransmitter acetylcholine (ACh) acts as an autocrine growth factor for human lung cancer. Several lines of evidence show that lung cancer cells express all of the proteins required for the uptake of choline (choline transporter 1, choline transporter-like proteins) synthesis of ACh (choline acetyltransferase, carnitine acetyltransferase), transport of ACh (vesicular acetylcholine transport, OCTs, OCTNs) and degradation of ACh (acetylcholinesterase, butyrylcholinesterase). The released ACh binds back to nicotinic (nAChRs) and muscarinic receptors on lung cancer cells to accelerate their proliferation, migration and invasion. Out of all components of the cholinergic pathway, the nAChR-signaling has been studied the most intensely. The reason for this trend is due to genome-wide data studies showing that nicotinic receptor subtypes are involved in lung cancer risk, the relationship between cigarette smoke and lung cancer risk as well as the rising popularity of electronic cigarettes considered by many as a "safe" alternative to smoking. There are a small number of articles which review the contribution of the other cholinergic proteins in the pathophysiology of lung cancer. The primary objective of this review article is to discuss the function of the acetylcholine-signaling proteins in the progression of lung cancer. The investigation of the role of cholinergic network in lung cancer will pave the way to novel molecular targets and drugs in this lethal malignancy.
Collapse
Affiliation(s)
- Jamie R Friedman
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Stephen D Richbart
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Justin C Merritt
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Kathleen C Brown
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Nicholas A Nolan
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Austin T Akers
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Jamie K Lau
- Biology Department, Center for the Sciences, Box 6931, Radford University, Radford, Virginia 24142
| | - Zachary R Robateau
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Sarah L Miles
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Piyali Dasgupta
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755.
| |
Collapse
|
26
|
Twayana KS, Ravanan P. Eukaryotic cell survival mechanisms: Disease relevance and therapeutic intervention. Life Sci 2018; 205:73-90. [PMID: 29730169 DOI: 10.1016/j.lfs.2018.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/17/2018] [Accepted: 05/01/2018] [Indexed: 01/10/2023]
Abstract
Cell responds to stress by activating various modes of stress responses which aim for minimal damage to cells and speedy recovery from the insults. However, unresolved stresses exceeding the tolerance limit lead to cell death (apoptosis, autophagy etc.) that helps to get rid of damaged cells and protect cell integrity. Furthermore, aberrant stress responses are the hallmarks of several pathophysiologies (neurodegeneration, metabolic diseases, cancer etc.). The catastrophic remodulation of stress responses is observed in cancer cells in favor of their uncontrolled growth. Whereas pro-survival stress responses redirected to death signaling provokes excessive cell death in neurodegeneration. Clear understanding of such mechanistic link to disease progression is required in order to modulate these processes for new therapeutic targets. The current review explains this with respect to novel drug discoveries and other breakthroughs in therapeutics.
Collapse
Affiliation(s)
- Krishna Sundar Twayana
- Apoptosis and Cell Survival Research Laboratory, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu-632014, India
| | - Palaniyandi Ravanan
- Apoptosis and Cell Survival Research Laboratory, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu-632014, India.
| |
Collapse
|
27
|
Vialinin A, an Edible Mushroom-Derived p-Terphenyl Antioxidant, Prevents VEGF-Induced Neovascularization In Vitro and In Vivo. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018. [PMID: 29541344 PMCID: PMC5818888 DOI: 10.1155/2018/1052102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Increased side toxicities and development of drug resistance are the major concern for the cancer chemotherapy using synthetic drugs. Therefore, identification of novel natural antioxidants with potential therapeutic efficacies is important. In the present study, we have examined how the antioxidant and anti-inflammatory activities of vialinin A, a p-terphenyl compound derived from Chinese edible mushroom T. terrestris and T. vialis, prevents human umbilical vascular endothelial cell (HUVEC) neovascularization in vitro and in vivo models. Pretreatment of HUVECs with vialinin A prevents vascular endothelial growth factor- (VEGF) induced HUVEC cell growth in a dose-dependent manner. Further, vialinin A also inhibits VEGF-induced migration as well as tube formation of HUVECs. Treatment of HUVECs prevents VEGF-induced generation of reactive oxygen species (ROS) and malondialdehyde (MDA) and also inhibits VEGF-induced NF-κB nuclear translocation as well as DNA-binding activity. The VEGF-induced release of various angiogenic cytokines and chemokines in HUVECs was also significantly blunted by vialinin A. Most importantly, in a mouse model of Matrigel plug assay, vialinin A prevents the formation of new blood vessels and the expression of CD31 and vWF. Thus, our results indicate a novel role of vialinin A in the prevention of neovascularization and suggest that anticancer effects of vialinin A could be mediated through its potent antioxidant and antiangiogenic properties.
Collapse
|
28
|
Gao P, Niu N, Wei T, Tozawa H, Chen X, Zhang C, Zhang J, Wada Y, Kapron CM, Liu J. The roles of signal transducer and activator of transcription factor 3 in tumor angiogenesis. Oncotarget 2017; 8:69139-69161. [PMID: 28978186 PMCID: PMC5620326 DOI: 10.18632/oncotarget.19932] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is the development of new blood vessels, which is required for tumor growth and metastasis. Signal transducer and activator of transcription factor 3 (STAT3) is a transcription factor that regulates a variety of cellular events including proliferation, differentiation and apoptosis. Previous studies revealed that activation of STAT3 promotes tumor angiogenesis. In this review, we described the activities of STAT3 signaling in different cell types involved in angiogenesis. Particularly, we elucidated the molecular mechanisms of STAT3-mediated gene regulation in angiogenic endothelial cells in response to external stimulations such as hypoxia and inflammation. The potential for STAT3 as a therapeutic target was also discussed. Overall, this review provides mechanistic insights for the roles of STAT3 signaling in tumor angiogenesis.
Collapse
Affiliation(s)
- Peng Gao
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Na Niu
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Tianshu Wei
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Hideto Tozawa
- The Research Center for Advanced Science and Technology, Isotope Science Center, The University of Tokyo, Meguro-ku, Tokyo, Japan
| | - Xiaocui Chen
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Caiqing Zhang
- Department of Respiratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Jiandong Zhang
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Youichiro Wada
- The Research Center for Advanced Science and Technology, Isotope Science Center, The University of Tokyo, Meguro-ku, Tokyo, Japan
| | - Carolyn M Kapron
- Department of Biology, Trent University, Peterborough, Ontario, Canada
| | - Ju Liu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| |
Collapse
|
29
|
Role of the unfolded protein response in tumor cell characteristics and cancer outcome. Curr Opin Oncol 2017; 29:41-47. [PMID: 27845970 DOI: 10.1097/cco.0000000000000339] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW In the present review, we discuss the possible role of the unfolded protein response (UPR) in the acquisition of tumor cell characteristics and in the prognosis of cancer outcome, which could assist and contribute to the development of more promising therapeutic strategies. RECENT FINDINGS Accumulating evidence supports the idea that alteration of endoplasmic reticulum proteostasis is a key player in cancer development and aggressiveness. Some UPR components were reported as independent prognostic biomarker. Recent evidence supports a relationship between the UPR activation status and prognosis of tumors. This may represent an interesting avenue for better characterization of carcinogenesis and tumor type. SUMMARY The contribution of the UPR to the characteristics of malignant tumors is complex and dependent on both intrinsic (e.g. oncogene addiction) and extrinsic (e.g. hypoxia) contexts. Through adaptation to severe microenvironmental conditions, UPR branches are generally a survival strategy for cancer cells, which are able to cope with this challenging context. We address the question of whether the activation status of the UPR is related to tumor properties and discuss the role of the UPR in the clinical context.
Collapse
|
30
|
Nunez Lopez YO, Garufi G, Seyhan AA. Altered levels of circulating cytokines and microRNAs in lean and obese individuals with prediabetes and type 2 diabetes. MOLECULAR BIOSYSTEMS 2017; 13:106-121. [PMID: 27869909 DOI: 10.1039/c6mb00596a] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Today obesity and type 2 diabetes (T2D) have both reached epidemic proportions. However, our current understanding of the primary mechanisms leading to these diseases is still limited due to the complex multifactorial nature of the underlying phenomena. We hypothesize that the levels of specific cytokines and miRNAs vary across the diabetes spectrum and unique signatures associated with them may serve as early biomarkers of the disease and provide insights into respective pathogenetic mechanisms. In this study, we measured the circulating levels of cytokines and microRNAs (miRNAs) in lean and obese humans with prediabetes (n = 21), T2D (n = 17), and healthy controls (n = 20) (ORIGINS trial, NCT02226640). Data were analyzed by fitting linear models adjusted for confounding variables (BMI, age, and gender in the diabetes context and age, gender, and diabetes status in the obesity context) and implementing nonparametric randomization-based tests for statistical inference. Group differences and correlations (r > 0.3) between variables with P < 0.05 were considered significant. False discovery rates (FDR) correcting for multiple testing were calculated using the Benjamini-Hochberg correction. We found a number of circulating cytokines and miRNAs deregulated in subjects with obesity, prediabetes, and T2D. Specifically, cytokines IL-6, IL-8, IL-10, IL-12, and SFRP4, as well as miRNAs miR-21, miR-24.1, miR-27a, miR-28-3p, miR-29b, miR-30d, miR-34a, miR-93, miR-126, miR-146a, miR-148, miR-150, miR-155, and miR-223, significantly changed across the diabetes spectrum, and were associated with measures of pancreatic islet β cell function and glycemic control, among others. Notably, SFRP4 was the only studied cytokine that was significantly associated with obesity, prediabetes, and T2D, which underscores the important role of this molecule during disease development and progression. Our data suggest that changes in circulating miRNAs and cytokines may have clinical utility as biomarkers of prediabetes.
Collapse
Affiliation(s)
- Yury O Nunez Lopez
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, 301 East Princeton St., Orlando, FL 32804, USA.
| | - Gabriella Garufi
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, 301 East Princeton St., Orlando, FL 32804, USA.
| | - Attila A Seyhan
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, 301 East Princeton St., Orlando, FL 32804, USA. and Sanford
- Burnham Medical Research Institute, Orlando, FL, USA and Massachusetts Institute of Technology, Chemical Engineering Department Cambridge, MA, USA
| |
Collapse
|
31
|
Poupard N, Badarou P, Fasani F, Groult H, Bridiau N, Sannier F, Bordenave-Juchereau S, Kieda C, Piot JM, Grillon C, Fruitier-Arnaudin I, Maugard T. Assessment of Heparanase-Mediated Angiogenesis Using Microvascular Endothelial Cells: Identification of λ-Carrageenan Derivative as a Potent Anti Angiogenic Agent. Mar Drugs 2017; 15:md15050134. [PMID: 28486399 PMCID: PMC5450540 DOI: 10.3390/md15050134] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 04/21/2017] [Accepted: 04/27/2017] [Indexed: 01/22/2023] Open
Abstract
Heparanase is overexpressed by tumor cells and degrades the extracellular matrix proteoglycans through cleavage of heparan sulfates (HS), allowing pro-angiogenic factor release and thus playing a key role in tumor angiogenesis and metastasis. Here we propose new HS analogs as potent heparanase inhibitors: Heparin as a positive control, Dextran Sulfate, λ-Carrageenan, and modified forms of them obtained by depolymerization associated to glycol splitting (RD-GS). After heparanase activity assessment, 11 kDa RD-GS-λ-Carrageenan emerged as the most effective heparanase inhibitor with an IC50 of 7.32 ng/mL compared to 10.7 ng/mL for the 16 kDa unfractionated heparin. The fractionated polysaccharides were then tested in a heparanase-rich medium-based in vitro model, mimicking tumor microenvironment, to determine their effect on microvascular endothelial cells (HSkMEC) angiogenesis. As a preliminary study, we identified that under hypoxic and nutrient poor conditions, MCF-7 cancer cells released much more mature heparanase in their supernatant than in normal conditions. Then a MatrigelTM assay using HSkMEC cultured under hypoxic conditions in the presence (or not) of this heparanase-rich supernatant was realized. Adding heparanase-rich media strongly enhanced angiogenic network formation with a production of twice more pseudo-vessels than with the control. When sulfated polysaccharides were tested in this angiogenesis assay, RD-GS-λ-Carrageenan was identified as a promising anti-angiogenic agent.
Collapse
Affiliation(s)
- Nicolas Poupard
- Université de la Rochelle, UMR CNRS 7266, LIENSs, Equipe Approches Moléculaires, Environnement-Santé, Avenue Michel Crépeau, 17000 La Rochelle, France.
| | - Pamela Badarou
- Centre de Biophysique Moléculaire, UPR CNRS 4301, 45071 Orléans, France.
| | - Fabienne Fasani
- Centre de Biophysique Moléculaire, UPR CNRS 4301, 45071 Orléans, France.
| | - Hugo Groult
- Université de la Rochelle, UMR CNRS 7266, LIENSs, Equipe Approches Moléculaires, Environnement-Santé, Avenue Michel Crépeau, 17000 La Rochelle, France.
| | - Nicolas Bridiau
- Université de la Rochelle, UMR CNRS 7266, LIENSs, Equipe Approches Moléculaires, Environnement-Santé, Avenue Michel Crépeau, 17000 La Rochelle, France.
| | - Frédéric Sannier
- Université de la Rochelle, UMR CNRS 7266, LIENSs, Equipe Approches Moléculaires, Environnement-Santé, Avenue Michel Crépeau, 17000 La Rochelle, France.
| | - Stéphanie Bordenave-Juchereau
- Université de la Rochelle, UMR CNRS 7266, LIENSs, Equipe Approches Moléculaires, Environnement-Santé, Avenue Michel Crépeau, 17000 La Rochelle, France.
| | - Claudine Kieda
- Centre de Biophysique Moléculaire, UPR CNRS 4301, 45071 Orléans, France.
| | - Jean-Marie Piot
- Université de la Rochelle, UMR CNRS 7266, LIENSs, Equipe Approches Moléculaires, Environnement-Santé, Avenue Michel Crépeau, 17000 La Rochelle, France.
| | - Catherine Grillon
- Centre de Biophysique Moléculaire, UPR CNRS 4301, 45071 Orléans, France.
| | - Ingrid Fruitier-Arnaudin
- Université de la Rochelle, UMR CNRS 7266, LIENSs, Equipe Approches Moléculaires, Environnement-Santé, Avenue Michel Crépeau, 17000 La Rochelle, France.
| | - Thierry Maugard
- Université de la Rochelle, UMR CNRS 7266, LIENSs, Equipe Approches Moléculaires, Environnement-Santé, Avenue Michel Crépeau, 17000 La Rochelle, France.
| |
Collapse
|
32
|
Anti-tumor activity of wogonin, an extract from Scutellaria baicalensis, through regulating different signaling pathways. Chin J Nat Med 2017; 15:15-40. [PMID: 28259249 DOI: 10.1016/s1875-5364(17)30005-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/06/2016] [Indexed: 12/14/2022]
Abstract
Wogonin is a plant flavonoid compound extracted from Scutellaria baicalensis (Huang-Qin or Chinese skullcap) and has been studied thoroughly by many researchers till date for its anti-viral, anti-oxidant, anti-cancerous and neuro-protective properties. Numerous experiments conducted in vitro and in vivo have demonstrated wogonin's excellent tumor inhibitory properties. The anti-cancer mechanism of wogonin has been ascribed to modulation of various cell signaling pathways, including serine-threonine kinase Akt (also known as protein kinase B) and AMP-activated protein kinase (AMPK) pathways, p53-dependent/independent apoptosis, and inhibition of telomerase activity. Furthermore, wogonin also decreases DNA adduct formation with a carcinogenic compound 2-Aminofluorene and inhibits growth of drug resistant malignant cells and their migration and metastasis, without any side effects. Recently, newly synthesized wogonin derivatives have been developed with impressive anti-tumor activity. This review is the succinct appraisal of the pertinent articles on the mechanisms of anti-tumor properties of wogonin. We also summarize the potential of wogonin and its derivatives used alone or as an adjunct therapy for cancer treatment. Furthermore, pharmacokinetics and side effects of wogonin and its analogues have also been discussed.
Collapse
|
33
|
Khan MA, Srivastava SK, Bhardwaj A, Singh S, Arora S, Zubair H, Carter JE, Singh AP. Gemcitabine triggers angiogenesis-promoting molecular signals in pancreatic cancer cells: Therapeutic implications. Oncotarget 2016; 6:39140-50. [PMID: 25970774 PMCID: PMC4770762 DOI: 10.18632/oncotarget.3784] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/10/2015] [Indexed: 12/19/2022] Open
Abstract
Pancreatic tumor microenvironment (TME) is characterized by poor tumor-vasculature and extensive desmoplasia that together contribute to poor response to chemotherapy. It was recently shown that targeting of TME to inhibit desmoplasiatic reaction in a preclinical model resulted in increased microvessel-density and intratumoral drug concentration, leading to improved therapeutic response. This approach; however, failed to generate a favorable response in clinical trial. In that regard, we have previously demonstrated a role of gemcitabine-induced CXCR4 signaling as a counter-defense mechanism, which also promoted invasiveness of pancreatic cancer (PC) cells. Here, we investigated the effect of gemcitabine on endothelial cell phenotype. Gemcitabine-treatment of human-umbilical-vein-endothelial-cells (HUVECs) did not promote the growth of HUVECs; however, it was induced when treated with conditioned media from gemcitabine-treated (Gem-CM) PC cells due to increased cell-cycle progression and apoptotic-resistance. Moreover, treatment of HUVECs with Gem-CM resulted in capillary-like structure (CLS) formation and promoted their ability to migrate and invade through extracellular-matrix. Gemcitabine-treatment of PC cells induced expression of various growth factors/cytokines, including IL-8, which exhibited greatest upregulation. Further, IL-8 depletion in Gem-CM diminished its potency to promote angiogenic phenotypes. Together, these findings suggest an indirect effect of gemcitabine on angiogenesis, which, in light of our previous observations, may hold important clinical significance.
Collapse
Affiliation(s)
- Mohammad Aslam Khan
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Sanjeev K Srivastava
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Arun Bhardwaj
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Seema Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Sumit Arora
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Haseeb Zubair
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - James E Carter
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Ajay P Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA.,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
34
|
Yang T, Yao Q, Cao F, Liu Q, Liu B, Wang XH. Silver nanoparticles inhibit the function of hypoxia-inducible factor-1 and target genes: insight into the cytotoxicity and antiangiogenesis. Int J Nanomedicine 2016; 11:6679-6692. [PMID: 27994464 PMCID: PMC5154724 DOI: 10.2147/ijn.s109695] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a transcription factor that is activated upon exposure to hypoxic stress. It modulates a number of cellular responses including proliferation, apoptosis, angiogenesis, and metabolism by activating a panel of target genes in response to hypoxia. The HIF-1 level is often upregulated in the hypoxic microenvironment of solid tumors, which contributes to cancer treatment failure. Here we report that silver nanoparticles (AgNPs), which are widely used as an antimicrobial agent, are an effective inhibitor of HIF-1. AgNPs inhibited the activation of a HIF-dependent reporter construct after the cells were exposed to hypoxic conditions or treated with cobalt chloride, a hypoxia mimetic agent. The AgNPs also interfered with the accumulation of HIF-1α protein and the induction of the endogenous HIF target genes, VEGF-A and GLUT1. Since both HIF-1 and vascular endothelial growth factor-A play an important role in angiogenesis, AgNPs also inhibited angiogenesis in vitro. Our data reveal a new mechanism of how AgNPs act on cellular function, that is, they disrupt HIF signaling pathway. This finding provides a novel insight into how AgNPs can inhibit cancer cell growth and angiogenesis.
Collapse
Affiliation(s)
- Tieshan Yang
- Laboratory for Biomedical Photonics, Institute of Laser Engineering, Beijing University of Technology
| | - Qian Yao
- Laboratory for Biomedical Photonics, Institute of Laser Engineering, Beijing University of Technology
| | - Fei Cao
- Laboratory for Biomedical Photonics, Institute of Laser Engineering, Beijing University of Technology
| | - Qianqian Liu
- Laboratory for Biomedical Photonics, Institute of Laser Engineering, Beijing University of Technology
| | - Binlei Liu
- Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Xiu-Hong Wang
- Laboratory for Biomedical Photonics, Institute of Laser Engineering, Beijing University of Technology
| |
Collapse
|
35
|
Vilsmaier T, Rack B, Janni W, Jeschke U, Weissenbacher T. Angiogenic cytokines and their influence on circulating tumour cells in sera of patients with the primary diagnosis of breast cancer before treatment. BMC Cancer 2016; 16:547. [PMID: 27464822 PMCID: PMC4964055 DOI: 10.1186/s12885-016-2612-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 07/25/2016] [Indexed: 12/05/2022] Open
Abstract
Background Circulating tumour cells (CTCs) have been found to be a prognostic marker for reduced disease free survival, breast cancer–specific survival, and overall survival before the start of systemic treatment. Methods A total of 200 patients’ sera were included in this study, 100 patients being CTC positive and 100 patients being CTC negative. Matching criteria were histo-pathological grading, lymph node metastasis, hormone receptor status, TNM classification and survived breast cancer patients vs. deceased tumor associated patients. A multi cytokine/chemokine array was used to screen the sera for the angiogenic markers. Results Statistical significant correlation was exposed for sFlt1 values in regard to the CTC-Status. CTC negative patients displayed increased sFlt1 expression opposed to CTC positive breast cancer patients. Furthermore, significant enhanced PIGF values were also disclosed in CTC negative patients compared to patients being CTC positive. Analyzing the living patient collective we found significant differences in sFlt1 and PlGF values in regard to CTC negative and CTC positive patients. Conclusion Both vascular markers showed enhanced expression in the CTC negative patient collective. To continue, the collective graded G2 showed significantly enhanced sFlt1 expressions amongst patients with no CTCs. Moreover, the patient collective with no lymph node metastasis and CTC negativity indicated statistically significant increased sFlt1 values. A functional interaction of sFlt1 and PlGF was found, suggesting that their overexpression in tumour cells inhibits CTCs entering the peripheral blood. Furthermore, in regard to CTC negativity, sFlt1 and PlGF values may potentially serve as predictive markers. Trial registration The TRN of this study is NCT02181101 and the date of registration was the 4th of June 2014. The study was retrospectively registered.
Collapse
Affiliation(s)
- Theresa Vilsmaier
- Department of Obstetrics and Gynecology, Ludwig-Maximilians-University of Munich, Maistrasse 11, 80337, Munich, Germany
| | - Brigitte Rack
- Department of Obstetrics and Gynecology, Ludwig-Maximilians-University of Munich, Maistrasse 11, 80337, Munich, Germany
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, University Hospital Ulm, Ulm, Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, Ludwig-Maximilians-University of Munich, Maistrasse 11, 80337, Munich, Germany.
| | - Tobias Weissenbacher
- Department of Obstetrics and Gynecology, Ludwig-Maximilians-University of Munich, Maistrasse 11, 80337, Munich, Germany
| | | |
Collapse
|
36
|
Epithelial derived CTGF promotes breast tumor progression via inducing EMT and collagen I fibers deposition. Oncotarget 2016; 6:25320-38. [PMID: 26318291 PMCID: PMC4694834 DOI: 10.18632/oncotarget.4659] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/17/2015] [Indexed: 12/21/2022] Open
Abstract
Interactions among tumor cells, stromal cells, and extracellular matrix compositions are mediated through cytokines during tumor progression. Our analysis of 132 known cytokines and growth factors in published clinical breast cohorts and our 84 patient-derived xenograft models revealed that the elevated connective tissue growth factor (CTGF) in tumor epithelial cells significantly correlated with poor clinical prognosis and outcomes. CTGF was able to induce tumor cell epithelial-mesenchymal transition (EMT), and promote stroma deposition of collagen I fibers to stimulate tumor growth and metastasis. This process was mediated through CTGF-tumor necrosis factor receptor I (TNFR1)-IκB autocrine signaling. Drug treatments targeting CTGF, TNFR1, and IκB signaling each prohibited the EMT and tumor progression.
Collapse
|
37
|
Achour O, Poupard N, Bridiau N, Bordenave Juchereau S, Sannier F, Piot JM, Fruitier Arnaudin I, Maugard T. Anti-heparanase activity of ultra-low-molecular-weight heparin produced by physicochemical depolymerization. Carbohydr Polym 2016; 135:316-23. [DOI: 10.1016/j.carbpol.2015.08.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 07/11/2015] [Accepted: 08/14/2015] [Indexed: 12/27/2022]
|
38
|
Vacchelli E, Aranda F, Bloy N, Buqué A, Cremer I, Eggermont A, Fridman WH, Fucikova J, Galon J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch-Immunostimulation with cytokines in cancer therapy. Oncoimmunology 2015; 5:e1115942. [PMID: 27057468 DOI: 10.1080/2162402x.2015.1115942] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 10/29/2015] [Indexed: 02/07/2023] Open
Abstract
During the past decade, great efforts have been dedicated to the development of clinically relevant interventions that would trigger potent (and hence potentially curative) anticancer immune responses. Indeed, developing neoplasms normally establish local and systemic immunosuppressive networks that inhibit tumor-targeting immune effector cells, be them natural or elicited by (immuno)therapy. One possible approach to boost anticancer immunity consists in the (generally systemic) administration of recombinant immunostimulatory cytokines. In a limited number of oncological indications, immunostimulatory cytokines mediate clinical activity as standalone immunotherapeutic interventions. Most often, however, immunostimulatory cytokines are employed as immunological adjuvants, i.e., to unleash the immunogenic potential of other immunotherapeutic agents, like tumor-targeting vaccines and checkpoint blockers. Here, we discuss recent preclinical and clinical advances in the use of some cytokines as immunostimulatory agents in oncological indications.
Collapse
Affiliation(s)
- Erika Vacchelli
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France
| | - Fernando Aranda
- Group of Immune receptors of the Innate and Adaptive System, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS)
| | - Norma Bloy
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France
| | - Aitziber Buqué
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France
| | - Isabelle Cremer
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 13, Center de Recherche des Cordeliers, Paris, France
| | | | - Wolf Hervé Fridman
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 13, Center de Recherche des Cordeliers, Paris, France
| | - Jitka Fucikova
- Sotio, Prague, Czech Republic; Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Jérôme Galon
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Laboratory of Integrative Cancer Immunology, Center de Recherche des Cordeliers, Paris, France
| | - Radek Spisek
- Sotio, Prague, Czech Republic; Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1015, CICBT507, Villejuif, France
| | - Guido Kroemer
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France; Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
39
|
de Souza DA, Borges AC, Santana AC, Oliver C, Jamur MC. Mast Cell Proteases 6 and 7 Stimulate Angiogenesis by Inducing Endothelial Cells to Release Angiogenic Factors. PLoS One 2015; 10:e0144081. [PMID: 26633538 PMCID: PMC4669151 DOI: 10.1371/journal.pone.0144081] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/12/2015] [Indexed: 12/11/2022] Open
Abstract
Mast cell proteases are thought to be involved with tumor progression and neo-vascularization. However, their exact role is still unclear. The present study was undertaken to further elucidate the function of specific subtypes of recombinant mouse mast cell proteases (rmMCP-6 and 7) in neo-vascularization. SVEC4-10 cells were cultured on Geltrex® with either rmMCP-6 or 7 and tube formation was analyzed by fluorescence microscopy and scanning electron microscopy. Additionally, the capacity of these proteases to induce the release of angiogenic factors and pro and anti-angiogenic proteins was analyzed. Both rmMCP-6 and 7 were able to stimulate tube formation. Scanning electron microscopy showed that incubation with the proteases induced SVEC4-10 cells to invade the gel matrix. However, the expression and activity of metalloproteases were not altered by incubation with the mast cell proteases. Furthermore, rmMCP-6 and rmMCP-7 were able to induce the differential release of angiogenic factors from the SVEC4-10 cells. rmMCP-7 was more efficient in stimulating tube formation and release of angiogenic factors than rmMCP-6. These results suggest that the subtypes of proteases released by mast cells may influence endothelial cells during in vivo neo-vascularization.
Collapse
Affiliation(s)
- Devandir Antonio de Souza
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Antonio Carlos Borges
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ana Carolina Santana
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Constance Oliver
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Maria Célia Jamur
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
- * E-mail:
| |
Collapse
|
40
|
Skin rejuvenation with non-invasive pulsed electric fields. Sci Rep 2015; 5:10187. [PMID: 25965851 PMCID: PMC4428072 DOI: 10.1038/srep10187] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/19/2015] [Indexed: 01/09/2023] Open
Abstract
Degenerative skin diseases affect one third of individuals over the age of sixty. Current therapies use various physical and chemical methods to rejuvenate skin; but since the therapies affect many tissue components including cells and extracellular matrix, they may also induce significant side effects, such as scarring. Here we report on a new, non-invasive, non-thermal technique to rejuvenate skin with pulsed electric fields. The fields destroy cells while simultaneously completely preserving the extracellular matrix architecture and releasing multiple growth factors locally that induce new cells and tissue growth. We have identified the specific pulsed electric field parameters in rats that lead to prominent proliferation of the epidermis, formation of microvasculature, and secretion of new collagen at treated areas without scarring. Our results suggest that pulsed electric fields can improve skin function and thus can potentially serve as a novel non-invasive skin therapy for multiple degenerative skin diseases.
Collapse
|
41
|
Abstract
Recent data indicate that hepatic angiogenesis, regardless of the etiology, takes place in chronic liver diseases (CLDs) that are characterized by inflammation and progressive fibrosis. Because anti-angiogenic therapy has been found to be efficient in the prevention of fibrosis in experimental models of CLDs, it is suggested that blocking angiogenesis could be a promising therapeutic option in patients with advanced fibrosis. Consequently, efforts are being directed to revealing the mechanisms involved in angiogenesis during the progression of liver fibrosis. Literature evidences indicate that hepatic angiogenesis and fibrosis are closely related in both clinical and experimental conditions. Hypoxia is a major inducer of angiogenesis together with inflammation and hepatic stellate cells. These profibrogenic cells stand at the intersection between inflammation, angiogenesis and fibrosis and play also a pivotal role in angiogenesis. This review mainly focuses to give a clear view on the relevant features that communicate angiogenesis with progression of fibrosis in CLDs towards the-end point of cirrhosis that may be translated into future therapies. The pathogenesis of hepatic angiogenesis associated with portal hypertension, viral hepatitis, non-alcoholic fatty liver disease and alcoholic liver disease are also discussed to emphasize the various mechanisms involved in angiogenesis during liver fibrogenesis.
Collapse
Affiliation(s)
- Gülsüm Özlem Elpek
- Gülsüm Özlem Elpek, Department of Pathology, Akdeniz University Medical School, 07070 Antalya, Turkey
| |
Collapse
|
42
|
Vassilakopoulou M, Psyrri A, Argiris A. Targeting angiogenesis in head and neck cancer. Oral Oncol 2015; 51:409-15. [PMID: 25680863 DOI: 10.1016/j.oraloncology.2015.01.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 01/06/2015] [Accepted: 01/08/2015] [Indexed: 10/24/2022]
Abstract
Angiogenesis is a crucial step in tumor growth and metastasis. Head and neck squamous cell carcinomas (HNSCC) highly express angiogenesis factors, such as vascular endothelial growth factor (VEGF), which are associated with patient prognosis. Antiangiogenesis agents can potentially modulate tumor microenvironment and induce radiosensitivity and chemosensitivity. In this review, we discuss the molecular mechanisms underlying angiogenesis involved in HNSCC, preclinical data with antiangiogenesis agents as well as potential predictive biomarkers. We also review novel therapies under investigation and summarize the results of clinical trials using antiangiogenesis agents alone or in combination with conventional therapies in HNSCC.
Collapse
Affiliation(s)
| | - Amanda Psyrri
- Attikon Hospital and Medical School of Athens, Athens, Greece
| | - Athanassios Argiris
- Hygeia Hospital, Athens, Greece; University of Texas Health Science Center at San Antonio, TX, USA.
| |
Collapse
|
43
|
Wynendaele E, Verbeke F, D'Hondt M, Hendrix A, Van De Wiele C, Burvenich C, Peremans K, De Wever O, Bracke M, De Spiegeleer B. Crosstalk between the microbiome and cancer cells by quorum sensing peptides. Peptides 2015; 64:40-8. [PMID: 25559405 DOI: 10.1016/j.peptides.2014.12.009] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 12/24/2014] [Accepted: 12/24/2014] [Indexed: 12/21/2022]
Abstract
To date, the precise role of the human microbiome in health and disease states remains largely undefined. Complex and selective crosstalk systems between the microbiome and mammalian cells are also not yet reported. Research up till now mainly focused on bacterial synthesis of virulence factors, reactive oxygen/nitrogen species (ROS/RNS) and hydrogen sulphide, as well as on the activation of exogenous mutagen precursors by intestinal bacteria. We discovered that certain quorum sensing peptides, produced by bacteria, interact with mammalian cells, in casu cancer cells: Phr0662 (Bacillus sp.), EntF-metabolite (Enterococcus faecium) and EDF-derived (Escherichia coli) peptides initiate HCT-8/E11 colon cancer cell invasion, with Phr0662 also promoting angiogenesis. Our findings thus indicate that the human microbiome, through their quorum sensing peptides, may be one of the factors responsible for cancer metastasis.
Collapse
Affiliation(s)
- Evelien Wynendaele
- Drug Quality and Registration (DruQuaR) Group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent B-9000, Belgium
| | - Frederick Verbeke
- Drug Quality and Registration (DruQuaR) Group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent B-9000, Belgium
| | - Matthias D'Hondt
- Drug Quality and Registration (DruQuaR) Group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent B-9000, Belgium
| | - An Hendrix
- Department of Radiation Oncology and Experimental Cancer Research, Faculty of Medicine and Health Sciences, Ghent University Hospital, De Pintelaan 185, Ghent B-9000, Belgium
| | - Christophe Van De Wiele
- Department of Radiology and Nuclear Medicine, Faculty of Medicine and Health Sciences, Ghent University Hospital, De Pintelaan 185, Ghent B-9000, Belgium
| | - Christian Burvenich
- Comparative Physiology and Biometrics, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, Merelbeke B-9820, Belgium
| | - Kathelijne Peremans
- Department of Medical Imaging, Medicine and Clinical Biology of Small Animals, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, Merelbeke B-9820, Belgium
| | - Olivier De Wever
- Department of Radiation Oncology and Experimental Cancer Research, Faculty of Medicine and Health Sciences, Ghent University Hospital, De Pintelaan 185, Ghent B-9000, Belgium
| | - Marc Bracke
- Department of Radiation Oncology and Experimental Cancer Research, Faculty of Medicine and Health Sciences, Ghent University Hospital, De Pintelaan 185, Ghent B-9000, Belgium
| | - Bart De Spiegeleer
- Drug Quality and Registration (DruQuaR) Group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent B-9000, Belgium.
| |
Collapse
|
44
|
Cooley-Andrade O, Jothidas A, Goh WX, Connor DE, Parsi K. Low-concentration detergent sclerosants stimulate white blood cells and release proinflammatory and proangiogenic cytokines in vitro. J Vasc Surg Venous Lymphat Disord 2014; 2:433-40. [PMID: 26993550 DOI: 10.1016/j.jvsv.2014.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 02/17/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE The objective of this study was to investigate the effects of the detergent sclerosants sodium tetradecyl sulfate (STS) and polidocanol (POL) on the release of proinflammatory and angiogenic mediators from circulating blood cells. METHODS Whole blood, platelet-rich plasma, platelet-poor plasma, and suspensions of polymorphonuclear cells (PMNCs) and mononuclear cells (MNCs) were incubated with varying concentrations of sclerosants. Enzyme-linked immunosorbent assay was used to detect the release of interferon-γ (IFN-γ); tumor necrosis factor-α (TNF-α); interleukins (IL) 1α, 1β, 6, 8, and 17; vascular endothelial growth factor (VEGF); and basic fibroblast growth factor. Leukocyte activation was assessed by flow cytometry. RESULTS IFN-γ and TNF-α were released from STS-stimulated PMNCs at high (387 ng/mL; P < .01) and modest (232 ng/mL; P < .05) quantities, respectively. Both sclerosants induced a weak response in MNCs, releasing minor quantities of IL-1α (STS, 18.2 ng/mL; POL, 14.0 ng/mL), IFN-γ (STS, 41.7 ng/mL; POL, 27.7 ng/mL), and TNF-α (STS, 29.7 ng/mL; POL, 14.0 ng/mL). POL at a wide range of concentrations and in all sample types tested stimulated the release of VEGF, whereas the effect of STS was limited to low concentrations and not detected in MNCs. Both agents stimulated moderate release of IL-8 from PMNCs (STS, 223.3 ng/mL, P < .001; POL, 84.23 ng/mL, P < .05). Neither agent induced a significant rise in basic fibroblast growth factor, IL-6, or IL-17. STS at low concentrations increased the expression of CD11b in both PMNCs and MNCs, suggestive of cellular activation. The activating effect of POL was limited to MNCs. CONCLUSIONS STS demonstrated proinflammatory activity mediated primarily by IFN-γ released from PMNCs and MNCs. POL had a weak proinflammatory effect limited to MNCs. Both sclerosants (POL > STS) induced a proangiogenic response mediated by VEGF.
Collapse
Affiliation(s)
- Osvaldo Cooley-Andrade
- Dermatology, Phlebology and Fluid Mechanics Research Laboratory, St. Vincent's Centre for Applied Medical Research, St. Vincent's Hospital, Sydney, New South Wales, Australia; University of New South Wales, Sydney, New South Wales, Australia
| | - Arunn Jothidas
- Dermatology, Phlebology and Fluid Mechanics Research Laboratory, St. Vincent's Centre for Applied Medical Research, St. Vincent's Hospital, Sydney, New South Wales, Australia; University of New South Wales, Sydney, New South Wales, Australia
| | - Wan Xian Goh
- Dermatology, Phlebology and Fluid Mechanics Research Laboratory, St. Vincent's Centre for Applied Medical Research, St. Vincent's Hospital, Sydney, New South Wales, Australia; University of New South Wales, Sydney, New South Wales, Australia
| | - David Ewan Connor
- Dermatology, Phlebology and Fluid Mechanics Research Laboratory, St. Vincent's Centre for Applied Medical Research, St. Vincent's Hospital, Sydney, New South Wales, Australia; University of New South Wales, Sydney, New South Wales, Australia
| | - Kurosh Parsi
- Dermatology, Phlebology and Fluid Mechanics Research Laboratory, St. Vincent's Centre for Applied Medical Research, St. Vincent's Hospital, Sydney, New South Wales, Australia; University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
45
|
In vitro inhibitory effect of crab shell extract on human umbilical vein endothelial cell. In Vitro Cell Dev Biol Anim 2014; 51:36-41. [DOI: 10.1007/s11626-014-9810-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 08/13/2014] [Indexed: 01/14/2023]
|
46
|
Gacche RN, Meshram RJ. Angiogenic factors as potential drug target: Efficacy and limitations of anti-angiogenic therapy. Biochim Biophys Acta Rev Cancer 2014; 1846:161-79. [DOI: 10.1016/j.bbcan.2014.05.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/05/2014] [Accepted: 05/07/2014] [Indexed: 12/17/2022]
|
47
|
Vacchelli E, Aranda F, Obrist F, Eggermont A, Galon J, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Immunostimulatory cytokines in cancer therapy. Oncoimmunology 2014; 3:e29030. [PMID: 25083328 PMCID: PMC4091551 DOI: 10.4161/onci.29030] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 04/26/2014] [Indexed: 12/11/2022] Open
Abstract
Tumor-targeting immune responses provide a significant contribution to (when they do not entirely account for) the clinical activity of diverse antineoplastic regimens, encompassing not only a large panel of immunotherapeutic strategies but also conventional cytotoxic molecules, targeted anticancer agents and irradiation. In line with this notion, several approaches have been devised to elicit novel or boost existing anticancer immune responses, including the administration of immunomodulatory cytokines. Such a relatively unspecific intervention suffices to mediate clinical effects in (at least a subset of) patients bearing particularly immunogenic tumors, like melanoma and renal cell carcinoma. More often, however, immunostimulatory cytokines are administered to boost the immunogenic potential of other agents, including (but not limited to) immune checkpoint-blocking antibodies, anticancer vaccines, oncolytic viruses and immunogenic chemotherapeutics. Here, we summarize the latest advances in the clinical development of recombinant cytokines as an immunomodulatory intervention for cancer therapy.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy; Villejuif, France ; INSERM, UMRS1138; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
| | - Fernando Aranda
- Gustave Roussy; Villejuif, France ; INSERM, UMRS1138; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
| | - Florine Obrist
- Gustave Roussy; Villejuif, France ; INSERM, UMRS1138; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
| | | | - Jérôme Galon
- INSERM, UMRS1138; Paris, France ; Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; Laboratory of Integrative Cancer Immunology, Centre de Recherche des Cordeliers; Paris, France
| | - Isabelle Cremer
- INSERM, UMRS1138; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France ; INSERM, U1015, CICBT507; Villejuif, France
| | - Guido Kroemer
- INSERM, UMRS1138; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France ; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France ; Metabolomics and Cell Biology Platforms, Gustave Roussy; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France
| |
Collapse
|
48
|
Signal transduction in tumor angiogenesis. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
49
|
Fan M, Zhang J, Wang Z, Wang B, Zhang Q, Zheng C, Li T, Ni C, Wu Z, Shao Z, Hu X. Phosphorylated VEGFR2 and hypertension: potential biomarkers to indicate VEGF-dependency of advanced breast cancer in anti-angiogenic therapy. Breast Cancer Res Treat 2013; 143:141-51. [PMID: 24292957 DOI: 10.1007/s10549-013-2793-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 11/23/2013] [Indexed: 12/27/2022]
Abstract
The efficacy of anti-VEGF agents probably lies on VEGF-dependency. Apatinib, a specific tyrosine kinase inhibitor that targets VEGF receptor 2, was assessed in patients with advanced breast cancer (ABC) (ClinicalTrials.gov NCT01176669 and NCT01653561). This substudy was to explore the potential biomarkers for VEGF-dependency in apatinib-treated breast cancer. Eighty pretreated patients received apatinib 750 or 500 mg/day orally in 4-week cycles. Circulating biomarkers were measured using a multiplex assay, and tissue biomarkers were identified with immunostaining. Baseline characteristics and adverse events (AEs) were included in the analysis. Statistical confirmation of independent predictive factors for anti-tumor efficacy was performed using Cox and Logistic regression models. Median progression-free survival (PFS) was 3.8 months, and overall survival (OS) was 10.6 months, with 17.5 % of objective response rate. Prominent AEs (≥60 %) were hypertension, hand-foot skin reaction (HFSR), and proteinuria. Higher tumor phosphorylated VEGFR2 (p-VEGFR2) expressions (P = 0.001), higher baseline serum soluble VEGFR2 (P = 0.031), hypertension (P = 0.011), and HFSR (P = 0.018) were significantly related to longer PFS, whereas hypertension (P = 0.002) and HFSR (P = 0.001) were also related to OS. Based on multivariate analysis, only p-VEGFR2 (adjusted HR, 0.40; P = 0.013) and hypertension (adjusted HR, 0.58; P = 0.038) were independent predictive factors for both PFS and clinical benefit rate. Apatinib had substantial antitumor activity in ABC and manageable toxicity. p-VEGFR2 and hypertension may be surrogate predictors of VEGF-dependency of breast cancer, which may identify an anti-angiogenesis sensitive population.
Collapse
Affiliation(s)
- Minhao Fan
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ge H, Tian P, Guan L, Yin X, Liu H, Xiao N, Xiong Y, Luo X, Sun Y, Qi D, Ni S, Liu P. A C-terminal fragment BIGH3 protein with an RGDRGD motif inhibits corneal neovascularization in vitro and in vivo. Exp Eye Res 2013; 112:10-20. [DOI: 10.1016/j.exer.2013.03.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Revised: 02/08/2013] [Accepted: 03/20/2013] [Indexed: 01/25/2023]
|