1
|
Shi Z, Kuai M, Li B, Akowuah CF, Wang Z, Pan Y, Tang M, Yang X, Lü P. The role of VEGF in Cancer angiogenesis and tumorigenesis: Insights for anti-VEGF therapy. Cytokine 2025; 189:156908. [PMID: 40049050 DOI: 10.1016/j.cyto.2025.156908] [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: 01/07/2025] [Revised: 02/16/2025] [Accepted: 02/28/2025] [Indexed: 03/18/2025]
Abstract
Vascular endothelial growth factor (VEGF) is a critical regulator of angiogenesis, playing a pivotal role in both physiological and pathological processes. It promotes the formation of new blood vessels and activates downstream signaling pathways that regulate endothelial cell function. This review highlights recent advancements in the understanding of VEGF's molecular structure and its isoforms, as well as their implications in disease progression. It also explores the mechanisms of VEGF inhibitors. While VEGF inhibitors show promise in the treatment of cancer and other diseases, their clinical use faces significant challenges, including drug resistance, side effects, and complex interactions with other signaling pathways. To address these challenges, future research should focus on: (i) enhancing the understanding of VEGF subtypes and their distinct roles in various diseases, supporting the development of personalized treatment strategies; (ii) developing combination therapies that integrate VEGF inhibitors with other targeted treatments to overcome resistance and improve efficacy; (iii) optimizing drug delivery systems to reduce off-target effects and enhance therapeutic outcomes. These approaches aim to improve the effectiveness and safety of VEGF-targeted therapies, offering new possibilities for the treatment of VEGF-related diseases.
Collapse
Affiliation(s)
- Zijun Shi
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Mengmeng Kuai
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Baohua Li
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | | | - Zhenyu Wang
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Ye Pan
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Min Tang
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Xiaoyue Yang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, China.
| | - Peng Lü
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China; Affiliated Hospital of Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
2
|
Lin QX, Song WW, Xie WX, Deng YT, Gong YN, Liu YR, Tian Y, Zhao WY, Tian L, Gu DN. Sequential treatment of anti-PD-L1 therapy prior to anti-VEGFR2 therapy contributes to more significant clinical benefits in non-small cell lung cancer. Neoplasia 2025; 59:101077. [PMID: 39561585 PMCID: PMC11617296 DOI: 10.1016/j.neo.2024.101077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024]
Abstract
OBJECTIVE Anti-angiogenic therapy and immune checkpoint blockade therapy are currently important treatments for non-small cell lung cancer. However, the combined use of the two therapies is controversial, and few studies have investigated the effects of different time sequences of the two therapies on treatment outcomes. METHODS The tumor-bearing mouse model was established and the mice were divided into four groups, including AA-ICB sequence group, ICB-AA sequence group, synchronization group and the control group. Immunohistochemistry was used to assess tumor microvessels and PD-L1 expression. Selected immune cell populations were evaluated using flow cytometry. Meta-analysis and clinical information were used to elucidate the clinical effects of administration sequence. RESULTS We found that anti-PD-L1 treatment followed by anti-VEGFR2 therapy exerts the best inhibitory effect on tumor growth. Different sequences of anti-angiogenic therapy and immune checkpoint blockade therapy resulted in different proportions of tumor microvessels and immune cell populations in the tumor microenvironment. We further revealed that the administration of anti-PD-L1 before anti-VEGFR brought more normalized tumor blood vessels and CD8+T cell infiltration and reduced immunosuppressive cells in the tumor microenvironment. Subsequent re-transplantation experiments confirmed the long-term benefits of this treatment strategy. The meta-analysis reinforced that immunotherapy prior to anti-angiogenic therapy or combination therapy have better therapeutic effects in advanced non-small cell lung cancer. CONCLUSION Our study demonstrated that the therapeutic effect of anti-angiogenic treatment after immune checkpoint therapy was superior to that of concurrent therapy, whereas anti-angiogenic therapy followed by immunotherapy did not bring more significant clinical benefits than independent monotherapy.
Collapse
Affiliation(s)
- Qiao-Xin Lin
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wen-Wen Song
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wen-Xia Xie
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi-Ting Deng
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan-Na Gong
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi-Ru Liu
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi Tian
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Ya Zhao
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Tian
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Dian-Na Gu
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
3
|
Eikanger MM, Sane S, Schraufnagel KS, Slunecka JL, Potts RA, Freeling J, Sereda G, Rasulev B, Brockstein RL, Emon MAB, Saif MTA, Rezvani K. Veratridine, a plant-derived alkaloid, suppresses the hyperactive Rictor-mTORC2 pathway: a new targeted therapy for primary and metastatic colorectal cancer. RESEARCH SQUARE 2024:rs.3.rs-5199838. [PMID: 39502780 PMCID: PMC11537347 DOI: 10.21203/rs.3.rs-5199838/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/14/2024]
Abstract
Despite considerable advances to improve colorectal cancer (CRC) survival over the last decade, therapeutic challenges remain due to the rapid metastatic dissemination of primary tumors and screening limitations. Meanwhile, the rise of CRC in younger adults (Early-onset CRC), commonly diagnosed with a metastatic form of the disease, shows the pressing need to develop more effective targeted therapies to decrease the high mortality rates associated with metastatic disease. Hyperactivation of the Rictor-mTORC2-AKT signaling pathway drives key metastatic players in diverse malignant tumors, including early- and late-onset colorectal cancer. Selective mTORC2 inhibitors are becoming a potential treatment strategy for CRC due to the therapeutic limitations of mTORC1 inhibitors. Veratridine (VTD), a lipid-soluble alkaloid extracted from Liliaceae plants, can transcriptionally increase UBXN2A, which induces 26S proteasomal degradation of the Rictor protein, a key member in the mTORC2 complex. Destabilization of Rictor protein by VTD decreases Akt phosphorylation on Ser473, which is responsible for metastatic signaling downstream of the mTORC2 pathway in diverse malignant tumors. VTD decreases the population of metastatic colon cancer stem cells and functions as an angiogenesis inhibitor. VTD effectively reduces the spheroid growth rate and restricts cell migration. Live cell migration and invasion assays alongside biomechanical-force-based experiments revealed that VTD suppresses colon cancer cell invasiveness and the ensuing risk of tumor metastasis. A CRC mouse model that mimics the natural stages of human sporadic CRC revealed that VTD treatment significantly decreases tumor growth in a UBXN2A-dependent manner. This study showed a novel mechanistic connection between a ubiquitin-like protein and mTORC2-dependent migration and invasion in CRC tumors. This study revealed the therapeutic benefit of selective inhibition of Rictor in CRC, particularly in tumors with a hyperactive Rictor-mTORC2 signaling pathway. Finally, this study opened a new platform for repurposing VTD, a supplemental anti-hypertension molecule, into an effective targeted therapy in CRC tumors.
Collapse
Affiliation(s)
| | - Sanam Sane
- University of South Dakota Sanford School of Medicine
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Sabit H, Arneth B, Abdel-Ghany S, Madyan EF, Ghaleb AH, Selvaraj P, Shin DM, Bommireddy R, Elhashash A. Beyond Cancer Cells: How the Tumor Microenvironment Drives Cancer Progression. Cells 2024; 13:1666. [PMID: 39404428 PMCID: PMC11475877 DOI: 10.3390/cells13191666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024] Open
Abstract
Liver cancer represents a substantial global health challenge, contributing significantly to worldwide morbidity and mortality. It has long been understood that tumors are not composed solely of cancerous cells, but also include a variety of normal cells within their structure. These tumor-associated normal cells encompass vascular endothelial cells, fibroblasts, and various inflammatory cells, including neutrophils, monocytes, macrophages, mast cells, eosinophils, and lymphocytes. Additionally, tumor cells engage in complex interactions with stromal cells and elements of the extracellular matrix (ECM). Initially, the components of what is now known as the tumor microenvironment (TME) were thought to be passive bystanders in the processes of tumor proliferation and local invasion. However, recent research has significantly advanced our understanding of the TME's active role in tumor growth and metastasis. Tumor progression is now known to be driven by an intricate imbalance of positive and negative regulatory signals, primarily influenced by specific growth factors produced by both inflammatory and neoplastic cells. This review article explores the latest developments and future directions in understanding how the TME modulates liver cancer, with the aim of informing the design of novel therapies that target critical components of the TME.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt; (H.S.); (E.F.M.)
| | - Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Philipps University Marburg, Baldinger Str., 35043 Marburg, Germany
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, Feulgenstr. 12, 35392 Giessen, Germany
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt;
| | - Engy F. Madyan
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt; (H.S.); (E.F.M.)
| | - Ashraf H. Ghaleb
- Department of Surgery, College of Medicine, Misr University for Science and Technology, Giza P.O. Box 77, Egypt;
- Department of Surgery, College of Medicine, Cairo University, Giza 12613, Egypt
| | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (P.S.); (R.B.)
| | - Dong M. Shin
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Ramireddy Bommireddy
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (P.S.); (R.B.)
| | - Ahmed Elhashash
- Department of Biology, Texas A&M University, 3258 TAMU I, College Station, TX 77843-3258, USA
| |
Collapse
|
5
|
Wang Z, Wang M, Guo J, Lu Y, Wei P. Adaptive resistance of tumor cells to anti-vascular endothelial growth factor therapy: A reversible phenomenon. Cytokine 2024; 180:156674. [PMID: 38852491 DOI: 10.1016/j.cyto.2024.156674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/06/2024] [Accepted: 06/05/2024] [Indexed: 06/11/2024]
Abstract
Vascular endothelial growth factor (VEGF) inhibition is an essential targeted strategy for malignant tumors, but its efficacy is severely constrained by drug resistance. The traditional view holds that the target of VEGF inhibition is endothelial cells, and thus compensatory angiogenesis is considered the main mechanism of drug resistance. In this study, we found that tumor cells themselves could develop acquired resistance to VEGF therapy, indicating an independent resistance mechanism apart from angiogenesis. Notably, this acquired resistance was temporary, disappearing completely four days after discontinuing exposure to the drug in vitro. Our findings suggest that tumor cells may also be targets of VEGF inhibition, and their response to treatment should not be overlooked in contributing to drug resistance.
Collapse
Affiliation(s)
- Zhiyong Wang
- Department of Immunology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Min Wang
- Department of Pharmaceutics, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Jinjin Guo
- Department of Immunology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Yanxin Lu
- Department of Immunology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Pei Wei
- Department of Immunology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China.
| |
Collapse
|
6
|
Grobbelaar C, Kgomo M, Mabeta P. Angiogenesis and Pancreatic Cancer: Novel Approaches to Overcome Treatment Resistance. Curr Cancer Drug Targets 2024; 24:1116-1127. [PMID: 38299403 DOI: 10.2174/0115680096284588240105051402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 02/02/2024]
Abstract
Pancreatic cancer (PCa) is acknowledged as a significant contributor to global cancer- related mortality and is widely recognized as one of the most challenging malignant diseases to treat. Pancreatic ductal adenocarcinoma (PDAC), which is the most common type of PCa, is highly aggressive and is mostly incurable. The poor prognosis of this neoplasm is exacerbated by the prevalence of angiogenic molecules, which contribute to stromal stiffness and immune escape. PDAC overexpresses various proangiogenic proteins, including vascular endothelial growth factor (VEGF)-A, and the levels of these molecules correlate with poor prognosis and treatment resistance. Moreover, VEGF-targeting anti-angiogenesis treatments are associated with the onset of resistance due to the development of hypoxia, which in turn induces the production of angiogenic molecules. Furthermore, excessive angiogenesis is one of the hallmarks of the second most common form of PCa, namely, pancreatic neuroendocrine tumor (PNET). In this review, the role of angiogenesis regulators in promoting disease progression in PCa, and the impact of these molecules on resistance to gemcitabine and various therapies against PCa are discussed. Finally, the use of anti-angiogenic agents in combination with chemotherapy and other targeted therapeutic molecules is discussed as a novel solution to overcome current treatment limitations in PCa.
Collapse
Affiliation(s)
- Craig Grobbelaar
- Department of Physiology, University of Pretoria, CNR Lynnwood Road and Roper Street, Hatfield, 0028, South Africa
| | - Mpho Kgomo
- Department of Internal Medicine, Faculty of Health Sciences, University of Pretoria, 9 Bophelo Road, Arcadia, CNR Lynnwood Road and Roper Street, Hatfield, 0028, South Africa
| | - Peace Mabeta
- Department of Physiology, University of Pretoria, CNR Lynnwood Road and Roper Street, Hatfield, 0028South Africa
| |
Collapse
|
7
|
Johnson BM, Johnson AM, Heim M, Buckley M, Mortimer B, Berry JL, Sewell-Loftin MK. Biomechanical stimulation promotes blood vessel growth despite VEGFR-2 inhibition. BMC Biol 2023; 21:290. [PMID: 38072992 PMCID: PMC10712065 DOI: 10.1186/s12915-023-01792-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Angiogenesis, or the growth of new vasculature from existing blood vessels, is widely considered a primary hallmark of cancer progression. When a tumor is small, diffusion is sufficient to receive essential nutrients; however, as the tumor grows, a vascular supply is needed to deliver oxygen and nutrients into the increasing mass. Several anti-angiogenic cancer therapies target VEGF and the receptor VEGFR-2, which are major promoters of blood vessel development. Unfortunately, many of these cancer treatments fail to completely stop angiogenesis in the tumor microenvironment (TME). Since these therapies focus on the biochemical activation of VEGFR-2 via VEGF ligand binding, we propose that mechanical cues, particularly those found in the TME, may be a source of VEGFR-2 activation that promotes growth of blood vessel networks even in the presence of VEGF and VEGFR-2 inhibitors. RESULTS In this paper, we analyzed phosphorylation patterns of VEGFR-2, particularly at Y1054/Y1059 and Y1214, stimulated via either VEGF or biomechanical stimulation in the form of tensile strains. Our results show prolonged and enhanced activation at both Y1054/Y1059 and Y1214 residues when endothelial cells were stimulated with strain, VEGF, or a combination of both. We also analyzed Src expression, which is downstream of VEGFR-2 and can be activated through strain or the presence of VEGF. Finally, we used fibrin gels and microfluidic devices as 3D microtissue models to simulate the TME. We determined that regions of mechanical strain promoted increased vessel growth, even with VEGFR-2 inhibition through SU5416. CONCLUSIONS Overall, understanding both the effects that biomechanical and biochemical stimuli have on VEGFR-2 activation and angiogenesis is an important factor in developing effective anti-angiogenic therapies. This paper shows that VEGFR-2 can be mechanically activated through strain, which likely contributes to increased angiogenesis in the TME. These proof-of-concept studies show that small molecular inhibitors of VEGFR-2 do not fully prevent angiogenesis in 3D TME models when mechanical strains are introduced.
Collapse
Affiliation(s)
- Bronte Miller Johnson
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6th Avenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, USA
| | - Allison McKenzie Johnson
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6th Avenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, USA
| | - Michael Heim
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6th Avenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, USA
| | - Molly Buckley
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6th Avenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, USA
| | - Bryan Mortimer
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Joel L Berry
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6th Avenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6th Avenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
8
|
Bhat BA, Saifi I, Khamjan NA, Hamdani SS, Algaissi A, Rashid S, Alshehri MM, Ganie SA, Lohani M, Abdelwahab SI, Dar SA. Exploring the tumor immune microenvironment in ovarian cancer: a way-out to the therapeutic roadmap. Expert Opin Ther Targets 2023; 27:841-860. [PMID: 37712621 DOI: 10.1080/14728222.2023.2259096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/21/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
INTRODUCTION Despite cancer treatment strides, mortality due to ovarian cancer remains high globally. While immunotherapy has proven effective in treating cancers with low cure rates, it has limitations. Growing evidence suggests that both tumoral and non-tumoral components of the tumor immune microenvironment (TIME) play a significant role in cancer growth. Therefore, developing novel and focused therapy for ovarian cancer is critical. Studies indicate that TIME is involved in developing ovarian cancer, particularly genome-, transcriptome-, and proteome-wide studies. As a result, TIME may present a prospective therapeutic target for ovarian cancer patients. AREAS COVERED We examined several TIME-targeting medicines and the connection between TIME and ovarian cancer. The key protagonists and events in the TIME and therapeutic strategies that explicitly target these events in ovarian cancer are discussed. EXPERT OPINION We highlighted various targeted therapies against TIME in ovarian cancer, including anti-angiogenesis therapies and immune checkpoint inhibitors. While these therapies are in their infancy, they have shown promise in controlling ovarian cancer progression. The use of 'omics' technology is helping in better understanding of TIME in ovarian cancer and potentially identifying new therapeutic targets. TIME-targeted strategies could account for an additional treatment strategy when treating ovarian cancer.
Collapse
Affiliation(s)
- Basharat Ahmad Bhat
- Department of Bioresources, Amar Singh College Campus, Cluster University, Srinagar, India
| | - Ifra Saifi
- Department of Botany, Chaudhary Charan Singh University, Meerut India
| | - Nizar A Khamjan
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Syed Suhail Hamdani
- Department of Bioresources, Amar Singh College Campus, Cluster University, Srinagar, India
| | - Abdullah Algaissi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
- Medical Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Safeena Rashid
- Department of Clinical Biochemistry, School of Biological Sciences, University of Kashmir, Srinagar, India
| | | | - Showkat Ahmad Ganie
- Department of Clinical Biochemistry, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Mohtashim Lohani
- Department of Emergency Medical Services, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | | | - Sajad Ahmad Dar
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
9
|
Szczerba K, Stokowa-Soltys K. What Is the Correlation between Preeclampsia and Cancer? The Important Role of Tachykinins and Transition Metal Ions. Pharmaceuticals (Basel) 2023; 16:366. [PMID: 36986466 PMCID: PMC10058266 DOI: 10.3390/ph16030366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/09/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Metal ions are irreplaceable in many biological processes. They are components of numerous metalloproteins and serve as cofactors or structural elements for enzymes. Interestingly, iron, copper and zinc play important roles in accelerating or preventing neoplastic cell transformation. Noteworthily, a lot of proliferative and invasive mechanisms are exploited by both malignant tumors and pregnancy. Cancer cells, as well as developing placenta cells, create a microenvironment supportive of immunologic privilege and angiogenesis. Therefore, pregnancy and cancer progression share many similarities. Moreover, during preeclampsia and cancer, significant changes in relevant trace element concentrations, tachykinin levels, expressions of neurokinin receptors, oxidative stress and angiogenic imbalance are observed. This sheds a new light on the role of metal ions and tachykinins in cancer progression and pregnancy, especially in preeclamptic women.
Collapse
Affiliation(s)
| | - Kamila Stokowa-Soltys
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland
| |
Collapse
|
10
|
Swamy K. Vascular normalization and immunotherapy: Spawning a virtuous cycle. Front Oncol 2022; 12:1002957. [PMID: 36276103 PMCID: PMC9582256 DOI: 10.3389/fonc.2022.1002957] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Anti-angiogenics, radiotherapy (especially stereotactic body radiotherapy, SBRT)/chemotherapy, and immunotherapy form a critical trimodal approach in modern cancer therapy. The normalization window, however short, is the beachhead for the strategic initiation of a decipherable disruption of cancer cells. This opening can be the opportunity for designing controlled stepwise cancer cell death (CCD) and immunological augmentation. The next step is to induce immunogenic cell death (ICD) through chemotherapy/radiotherapy concurrently with the facilitation of professional phagocytosis. Immunotherapy at this stage, when interstitial pressure decreases considerably, leads to the improved perfusion of oxygen with solutes and improved immune-friendly pH and is additionally expected to open up the tumor microenvironment (TME) for a “flood” of tumor-infiltrating lymphocytes. Furthermore, there would be enhanced interaction in “hot” nodules and the incorporation of immune reaction in “cold” nodules. Simultaneously, the added adjuvant-assisted neoantigen–immune cell interaction will likely set in a virtuous cycle of CCD induction followed by tumor cell-specific antigenic reaction boosting CCD, in turn promoting the normalization of the vasculature, completing the loop. There should be a conscious concern to protect the extracellular matrix (ECM), which will nurture the long-term immunological cross-talk to discourage dormancy, which is as essential as obtaining a complete response in imaging. The caveat is that the available therapies should be appropriately ranked during the start of the treatment since the initial administration is the most opportune period. A fast-paced development in the nanomedicine field is likely to assist in all the steps enumerated.
Collapse
|
11
|
Shao X, Hua S, Feng T, Ocansey DKW, Yin L. Hypoxia-Regulated Tumor-Derived Exosomes and Tumor Progression: A Focus on Immune Evasion. Int J Mol Sci 2022; 23:ijms231911789. [PMID: 36233088 PMCID: PMC9570495 DOI: 10.3390/ijms231911789] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/24/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor cells express a high quantity of exosomes packaged with unique cargos under hypoxia, an important characteristic feature in solid tumors. These hypoxic tumor-derived exosomes are, crucially, involved in the interaction of cancer cells with their microenvironment, facilitating not only immune evasion, but increased cell growth and survival, enhanced angiogenesis, epithelial–mesenchymal transition (EMT), therapeutic resistance, autophagy, pre-metastasis, and metastasis. This paper explores the tumor microenvironment (TME) remodeling effects of hypoxic tumor-derived exosome towards facilitating the tumor progression process, particularly, the modulatory role of these factors on tumor cell immune evasion through suppression of immune cells, expression of surface recognition molecules, and secretion of antitumor soluble factor. Tumor-expressed exosomes educate immune effector cells, including macrophages, monocytes, T cells, natural killer (NK) cells, dendritic cells (DCs), γδ T lymphocytes, regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), mast cells, and B cells, within the hypoxic TME through the release of factors that regulate their recruitment, phenotype, and function. Thus, both hypoxia and tumor-derived exosomes modulate immune cells, growth factors, cytokines, receptor molecules, and other soluble factors, which, together, collaborate to form the immune-suppressive milieu of the tumor environment. Exploring the contribution of exosomal cargos, such as RNAs and proteins, as indispensable players in the cross-talk within the hypoxic tumor microenvironmental provides a potential target for antitumor immunity or subverting immune evasion and enhancing tumor therapies.
Collapse
|
12
|
Hu X, Luo B, Qiu L, Chen S, Wu Q, Chen Q, Liu X, Ling C, Deng S, Yuan M, Hu P. Dezocine Has the Potential to Regulate the Clinical and Biological Features of Tumors. Drug Des Devel Ther 2022; 16:1121-1129. [PMID: 35478934 PMCID: PMC9035457 DOI: 10.2147/dddt.s356863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/09/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer is the second leading cause of death following ischemic heart disease in the world and the primary clinical, social and economic burden. Surgical resection is the main measure for the treatment of the vast majority of solid tumors. However, the recurrence and metastasis of tumors occur at different periods after surgery in many cases undergoing radical tumor surgery, which is the main cause of death of tumor patients. Moreover, tumor patients are prone to suffer from mental depression, which may increase the morbidity and mortality of tumors. Tumors have a series of clinical biological signs with the following five main features: postoperative pain and cancerous pain; suppression of antitumor immunity; angiogenesis in tumors; proliferation, growth and metastasis of tumors; and mental depression. Surgery is the first treatment in the majority of cancer patients with solid tumors. Opioids are required for anesthesia and postoperative analgesia. For cancerous pain control, patients undergo surgery, and their quality of life of is improved. However, traditional opioids, such as morphine, may inhibit antitumor immunity, induce vascular growth of tumors and promote the proliferation, invasion and migration of cancer cells, and traditional opioids can induce a risk of somatic dependence. However, studies have found that not all opioids share the effects of immunosuppression, tumor proliferation promotion and angiogenesis induction. Dezocine, a novel opioid with specific pharmacological mechanisms, has been demonstrated to regulate the five clinical and biological features of tumors. We reviewed the preclinical and clinical studies of dezocine on postoperative pain and cancer pain in tumor patients as well as the immune system, tumor angiogenesis, tumor proliferation, tumor growth, tumor metastasis and mental depression. We proposed that dezocine may be the best choice of opioids for anesthesia and analgesia in cancer patients.
Collapse
Affiliation(s)
- Xudong Hu
- Department of Anesthesiology, The Second People's Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Bing Luo
- Department of Surgery, The Second People's Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Lei Qiu
- Department of Surgery, The Second People's Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Shaosen Chen
- Department of Respiratory and Critical Care Medicine, The Second People's Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Qing Wu
- Department of Surgery, The Second People's Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Qingbiao Chen
- Department of Surgery, The Second People's Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Xingqing Liu
- Department of Anesthesiology, The Second People's Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Chen Ling
- Department of Anesthesiology, The Second People's Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Shuping Deng
- Department of Internal Medicine, Huanshi Hospital, People's Hospital of Chancheng District, Foshan, 528000, People's Republic of China
| | - Manjuan Yuan
- Department of Respiratory and Critical Care Medicine, The Second People's Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Peicun Hu
- Department of Respiratory and Critical Care Medicine, The Second People's Hospital of Foshan, Foshan, 528000, People's Republic of China
| |
Collapse
|
13
|
Miller B, Sewell-Loftin MK. Mechanoregulation of Vascular Endothelial Growth Factor Receptor 2 in Angiogenesis. Front Cardiovasc Med 2022; 8:804934. [PMID: 35087885 PMCID: PMC8787114 DOI: 10.3389/fcvm.2021.804934] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022] Open
Abstract
The endothelial cells that compose the vascular system in the body display a wide range of mechanotransductive behaviors and responses to biomechanical stimuli, which act in concert to control overall blood vessel structure and function. Such mechanosensitive activities allow blood vessels to constrict, dilate, grow, or remodel as needed during development as well as normal physiological functions, and the same processes can be dysregulated in various disease states. Mechanotransduction represents cellular responses to mechanical forces, translating such factors into chemical or electrical signals which alter the activation of various cell signaling pathways. Understanding how biomechanical forces drive vascular growth in healthy and diseased tissues could create new therapeutic strategies that would either enhance or halt these processes to assist with treatments of different diseases. In the cardiovascular system, new blood vessel formation from preexisting vasculature, in a process known as angiogenesis, is driven by vascular endothelial growth factor (VEGF) binding to VEGF receptor 2 (VEGFR-2) which promotes blood vessel development. However, physical forces such as shear stress, matrix stiffness, and interstitial flow are also major drivers and effectors of angiogenesis, and new research suggests that mechanical forces may regulate VEGFR-2 phosphorylation. In fact, VEGFR-2 activation has been linked to known mechanobiological agents including ERK/MAPK, c-Src, Rho/ROCK, and YAP/TAZ. In vascular disease states, endothelial cells can be subjected to altered mechanical stimuli which affect the pathways that control angiogenesis. Both normalizing and arresting angiogenesis associated with tumor growth have been strategies for anti-cancer treatments. In the field of regenerative medicine, harnessing biomechanical regulation of angiogenesis could enhance vascularization strategies for treating a variety of cardiovascular diseases, including ischemia or permit development of novel tissue engineering scaffolds. This review will focus on the impact of VEGFR-2 mechanosignaling in endothelial cells (ECs) and its interaction with other mechanotransductive pathways, as well as presenting a discussion on the relationship between VEGFR-2 activation and biomechanical forces in the extracellular matrix (ECM) that can help treat diseases with dysfunctional vascular growth.
Collapse
Affiliation(s)
- Bronte Miller
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
14
|
Senchukova MA. Issues of origin, morphology and clinical significance of tumor microvessels in gastric cancer. World J Gastroenterol 2021; 27:8262-8282. [PMID: 35068869 PMCID: PMC8717017 DOI: 10.3748/wjg.v27.i48.8262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/02/2021] [Accepted: 12/07/2021] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) remains a serious oncological problem, ranking third in the structure of mortality from malignant neoplasms. Improving treatment outcomes for this pathology largely depends on understanding the pathogenesis and biological characteristics of GC, including the identification and characterization of diagnostic, prognostic, predictive, and therapeutic biomarkers. It is known that the main cause of death from malignant neoplasms and GC, in particular, is tumor metastasis. Given that angiogenesis is a critical process for tumor growth and metastasis, it is now considered an important marker of disease prognosis and sensitivity to anticancer therapy. In the presented review, modern concepts of the mechanisms of tumor vessel formation and the peculiarities of their morphology are considered; data on numerous factors influencing the formation of tumor microvessels and their role in GC progression are summarized; and various approaches to the classification of tumor vessels, as well as the methods for assessing angiogenesis activity in a tumor, are highlighted. Here, results from studies on the prognostic and predictive significance of tumor microvessels in GC are also discussed, and a new classification of tumor microvessels in GC, based on their morphology and clinical significance, is proposed for consideration.
Collapse
Affiliation(s)
- Marina A Senchukova
- Department of Oncology, Orenburg State Medical University, Orenburg 460021, Russia
| |
Collapse
|
15
|
Bogusławska-Duch J, Ducher-Hanaka M, Zajkowska A, Czajka M, Małecki M. Therapeutic combination silencing VEGF and SOX10 increases the antiangiogenic effect in the mouse melanoma model B16-F10 - in vitro and in vivo studies. Postepy Dermatol Alergol 2021; 38:887-898. [PMID: 34849139 PMCID: PMC8610042 DOI: 10.5114/ada.2021.110461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/24/2020] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Gene therapy is an innovative form of treatment of genetic diseases, in which psiRNA molecules silencing specific genes are applied. AIM The study evaluated the anti-tumour effect of psiRNA silencing preparations of the vascular endothelial growth factor (VEGF) and Sry-related HMG-Box gene 10 (SOX10) on melanoma (B16-F10) by inhibiting angiogenesis. MATERIAL AND METHODS The preparations based on plasmid vectors psiRNA silencing the gene SOX10 and VEGF that form complexes with cationic lipid (psiRNA/carrier) have been developed. psiRNA preparations were tested on the mouse melanoma cell line B16-F10, both in vitro and in vivo. The silencing activity of transfected melanoma cells with the obtained psiRNA preparations was examined using the qPCR and Western blot methods. The anti-tumour activity of psiRNA preparations on melanoma tumour cells was then evaluated in a mouse in vivo model. RESULTS In vitro studies have shown that the B16-F10 cells efficiently transfect non-viral preparations - psiRNA: Lyovec (74-89%). Worth mentioning is the fact that silencing SOX10 in B16-F10 melanoma cells increases the expression of the COL18A1 gene (compared to the preparation inhibiting only VEGF), which codes the endostatin to stop angiogenesis. In vivo results show that the level of haemoglobin in tumours of mice treated with psiRNA formulations was over 6 times lower than controls and tumour mass was 60-80% lower. CONCLUSIONS The novel study proves that simultaneous inhibition of SOX10 and VEGF enhances the antiangiogenic action and thus contributes to a significant halt of disease development. In addition, these data expand knowledge about SOX10 regulation and functions.
Collapse
Affiliation(s)
| | | | - Agnieszka Zajkowska
- Department of Applied Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Milena Czajka
- Department of Applied Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Maciej Małecki
- Department of Applied Pharmacy, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
16
|
Jiang W, Hu JW, He XR, Jin WL, He XY. Statins: a repurposed drug to fight cancer. J Exp Clin Cancer Res 2021; 40:241. [PMID: 34303383 PMCID: PMC8306262 DOI: 10.1186/s13046-021-02041-2] [Citation(s) in RCA: 234] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
As competitive HMG-CoA reductase (HMGCR) inhibitors, statins not only reduce cholesterol and improve cardiovascular risk, but also exhibit pleiotropic effects that are independent of their lipid-lowering effects. Among them, the anti-cancer properties of statins have attracted much attention and indicated the potential of statins as repurposed drugs for the treatment of cancer. A large number of clinical and epidemiological studies have described the anticancer properties of statins, but the evidence for anticancer effectiveness of statins is inconsistent. It may be that certain molecular subtypes of cancer are more vulnerable to statin therapy than others. Whether statins have clinical anticancer effects is still an active area of research. Statins appear to enhance the efficacy and address the shortcomings associated with conventional cancer treatments, suggesting that statins should be considered in the context of combined therapies for cancer. Here, we present a comprehensive review of the potential of statins in anti-cancer treatments. We discuss the current understanding of the mechanisms underlying the anti-cancer properties of statins and their effects on different malignancies. We also provide recommendations for the design of future well-designed clinical trials of the anti-cancer efficacy of statins.
Collapse
Affiliation(s)
- Wen Jiang
- Department of General Surgery, The Affiliated Provincial Hospital of Anhui Medical University, Hefei, 230001, P. R. China
| | - Jin-Wei Hu
- Department of General Surgery, The Affiliated Provincial Hospital of Anhui Medical University, Hefei, 230001, P. R. China
| | - Xu-Ran He
- Department of Finance, The First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital), Hefei, 230001, P. R. China
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, P. R. China.
| | - Xin-Yang He
- Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital), Hefei, 230001, P. R. China.
| |
Collapse
|
17
|
Kadioglu O, Saeed MEM, Mahmoud N, Azawi S, Mrasek K, Liehr T, Efferth T. Identification of novel drug resistance mechanisms by genomic and transcriptomic profiling of glioblastoma cells with mutation-activated EGFR. Life Sci 2021; 284:119601. [PMID: 33991550 DOI: 10.1016/j.lfs.2021.119601] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 11/29/2022]
Abstract
AIMS Epidermal growth factor receptor (EGFR) is not only involved in carcinogenesis, but also in chemoresistance. We characterized U87.MGΔEGFR glioblastoma cells with constitutively active EGFR due to deletion at the ligand binding domain in terms of gene expression profiling and chromosomal aberrations. Wild-type U87.MG cells served as control. MATERIALS AND METHODS RNA sequencing and network analyses (Ingenuity Pathway Analysis) were performed to identify novel drug resistance mechanisms related to expression of mutation activated EGFR. Chromosomal aberrations were characterized by multicolor fluorescence in situ hybridization (mFISH) and array comparative genomic hybridization (aCGH). KEY FINDINGS U87.MGΔEGFR cells presented much more chromosomal aberrations, amplifications and deletions than wild-type U87.MG cells. Still, both cell lines were near-triploid. Numerous genes were overexpressed in U87.MGΔEGFR cells, some of which have been already linked to drug resistance. PXDN, which is associated with epithelial mesenchymal transition, was the most upregulated gene (901.8-fold). TENM1 was 331.6-fold upregulated, and it was previously reported to modulate neural development. EGFR-AS1 (161.2-fold upregulated) has been reported to increase the EGFR mRNA stability and its expression - in accordance with that of EGFR - was upregulated (85.5-fold). In addition to well-known resistance genes, numerous novel genes and genomic aberrations were identified. ANGPT2 upregulation and CPM downregulation were validated by Western blotting. SIGNIFICANCE Transcriptomics and genomics analyses in U87.MGΔEGFR cells unraveled a range of novel drug resistance mechanisms including apoptosis, DNA repair, ferroptosis, glutathione related gene activities, heat shock, oxidative stress, transcription factor activities, which may have important implications for future treatment strategies.
Collapse
Affiliation(s)
- Onat Kadioglu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Mohamed E M Saeed
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Nuha Mahmoud
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Shaymaa Azawi
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Jena, Germany
| | - Kristin Mrasek
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Jena, Germany
| | - Thomas Liehr
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Jena, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
18
|
Mandula JK, Rodriguez PC. Tumor-related stress regulates functional plasticity of MDSCs. Cell Immunol 2021; 363:104312. [PMID: 33652258 PMCID: PMC8026602 DOI: 10.1016/j.cellimm.2021.104312] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/15/2021] [Accepted: 01/29/2021] [Indexed: 12/15/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) impair protective anti-tumor immunity and remain major obstacles that stymie the effectiveness of promising cancer therapies. Diverse tumor-derived stressors galvanize the differentiation, intra-tumoral expansion, and immunomodulatory function of MDSCs. These tumor-associated 'axes of stress' underwrite the immunosuppressive programming of MDSCs in cancer and contribute to the phenotypic/functional heterogeneity that characterize tumor-MDSCs. This review discusses various tumor-associated axes of stress that direct MDSC development, accumulation, and immunosuppressive function, as well as current strategies aimed at overcoming the detrimental impact of MDSCs in cancer. To better understand the constellation of signals directing MDSC biology, we herein summarize the pivotal roles, signaling mediators, and effects of reactive oxygen/nitrogen species-related stress, chronic inflammatory stress, hypoxia-linked stress, endoplasmic reticulum stress, metabolic stress, and therapy-associated stress on MDSCs. Although therapeutic targeting of these processes remains mostly pre-clinical, intercepting signaling through the axes of stress could overcome MDSC-related immune suppression in tumor-bearing hosts.
Collapse
Affiliation(s)
- Jessica K Mandula
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
19
|
Liu JJ, Liu XY, Nie JP, Jia MQ, Yu Y, Qin N, Duan HQ. Discovery of Antimetastatic Chiral Ionone Alkaloid Derivatives Targeting HIF-1α/VEGF/VEGFR2 Pathway. ChemMedChem 2021; 16:2130-2145. [PMID: 33755334 DOI: 10.1002/cmdc.202100072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/07/2021] [Indexed: 11/10/2022]
Abstract
Novel chiral ionone alkaloid derivatives were synthesized and their antimetastatic effects were evaluated in human breast cancer cells using chemotaxis assay. Compared with positive control LY294002, a PI3 K inhibitor, derivatives 10 a, 11 a, 11 c, 11 g, 11 j, 11 k and 11 w exhibited significant inhibitory effects against cancer cell migration. Especially, the IC50 for compound 11 g was as low as 0.035±0.004 μM. Further investigations on compound 11 g revealed that it could exert inhibitory effects on the adhesion, migration and invasion of MDA-MB-231 cells. The mechanisms for the antitumor metastatic effects of 11 g might be through the inhibition of HIF-1α/VEGF/VEGFR2/Akt pathway, which suppressed the downstream signaling molecules, including Akt1/mTOR/p70S6K and Akt2/PKCζ/integrin β1 pathways. Taken together, chiral ionone alkaloid derivative 11 g has the potential to be developed into an antitumor metastatic agent for breast cancer.
Collapse
Affiliation(s)
- Jing-Jing Liu
- Tianjin Key Laboratory on Technologies Enabling Development Clinical Therapeutics and Diagnostics (Theragnostic), Tianjin Medical University, Tianjin, China
| | - Xin-Yao Liu
- Tianjin Key Laboratory on Technologies Enabling Development Clinical Therapeutics and Diagnostics (Theragnostic), Tianjin Medical University, Tianjin, China
| | - Jiang-Ping Nie
- Tianjin Key Laboratory on Technologies Enabling Development Clinical Therapeutics and Diagnostics (Theragnostic), Tianjin Medical University, Tianjin, China
| | - Mei-Qi Jia
- Tianjin Key Laboratory on Technologies Enabling Development Clinical Therapeutics and Diagnostics (Theragnostic), Tianjin Medical University, Tianjin, China
| | - Yang Yu
- Tianjin Key Laboratory on Technologies Enabling Development Clinical Therapeutics and Diagnostics (Theragnostic), Tianjin Medical University, Tianjin, China
| | - Nan Qin
- Tianjin Key Laboratory on Technologies Enabling Development Clinical Therapeutics and Diagnostics (Theragnostic), Tianjin Medical University, Tianjin, China
| | - Hong-Quan Duan
- Tianjin Key Laboratory on Technologies Enabling Development Clinical Therapeutics and Diagnostics (Theragnostic), Tianjin Medical University, Tianjin, China.,Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
20
|
Moosavi F, Giovannetti E, Peters GJ, Firuzi O. Combination of HGF/MET-targeting agents and other therapeutic strategies in cancer. Crit Rev Oncol Hematol 2021; 160:103234. [PMID: 33497758 DOI: 10.1016/j.critrevonc.2021.103234] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/29/2020] [Accepted: 01/16/2021] [Indexed: 02/06/2023] Open
Abstract
MET receptor has emerged as a druggable target across several human cancers. Agents targeting MET and its ligand hepatocyte growth factor (HGF) including small molecules such as crizotinib, tivantinib and cabozantinib or antibodies including rilotumumab and onartuzumab have proven their values in different tumors. Recently, capmatinib was approved for treatment of metastatic lung cancer with MET exon 14 skipping. In this review, we critically examine the current evidence on how HGF/MET combination therapies may take advantage of synergistic effects, overcome primary or acquired drug resistance, target tumor microenvironment, modulate drug metabolism or tackle pharmacokinetic issues. Preclinical and clinical studies on the combination of HGF/MET-targeted agents with conventional chemotherapeutics or molecularly targeted treatments (including EGFR, VEGFR, HER2, RAF/MEK, and PI3K/Akt targeting agents) and also the value of biomarkers are examined. Our deeper understanding of molecular mechanisms underlying successful pharmacological combinations is crucial to find the best personalized treatment regimens for cancer patients.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, the Netherlands; Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, the Netherlands; Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
21
|
The CXCL2/IL8/CXCR2 Pathway Is Relevant for Brain Tumor Malignancy and Endothelial Cell Function. Int J Mol Sci 2021; 22:ijms22052634. [PMID: 33807899 PMCID: PMC7961945 DOI: 10.3390/ijms22052634] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/30/2022] Open
Abstract
We aimed to evaluate the angiogenic capacity of CXCL2 and IL8 affecting human endothelial cells to clarify their potential role in glioblastoma (GBM) angiogenesis. Human GBM samples and controls were stained for proangiogenic factors. Survival curves and molecule correlations were obtained from the TCGA (The Cancer Genome Atlas) database. Moreover, proliferative, migratory and angiogenic activity of peripheral (HUVEC) and brain specific (HBMEC) primary human endothelial cells were investigated including blockage of CXCR2 signaling with SB225502. Gene expression analyses of angiogenic molecules from endothelial cells were performed. Overexpression of VEGF and CXCL2 was observed in GBM patients and associated with a survival disadvantage. Molecules of the VEGF pathway correlated but no relation for CXCR1/2 and CXCL2/IL8 was found. Interestingly, receptors of endothelial cells were not induced by addition of proangiogenic factors in vitro. Proliferation and migration of HUVEC were increased by VEGF, CXCL2 as well as IL8. Their sprouting was enhanced through VEGF and CXCL2, while IL8 showed no effect. In contrast, brain endothelial cells reacted to all proangiogenic molecules. Additionally, treatment with a CXCR2 antagonist led to reduced chemokinesis and sprouting of endothelial cells. We demonstrate the impact of CXCR2 signaling on endothelial cells supporting an impact of this pathway in angiogenesis of glioblastoma.
Collapse
|
22
|
Liu L, Xing L, Chen R, Zhang J, Huang Y, Huang L, Xie B, Ren X, Wang S, Kuang H, Lin X, Kumar A, Kim JK, Lee C, Li X. Mitogen-Inducible Gene 6 Inhibits Angiogenesis by Binding to SHC1 and Suppressing Its Phosphorylation. Front Cell Dev Biol 2021; 9:634242. [PMID: 33693003 PMCID: PMC7937727 DOI: 10.3389/fcell.2021.634242] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/05/2021] [Indexed: 12/17/2022] Open
Abstract
The mitogen-inducible gene 6 (MIG6) is an adaptor protein widely expressed in vascular endothelial cells. However, it remains unknown thus far whether it plays a role in angiogenesis. Here, using comprehensive in vitro and in vivo model systems, we unveil a potent anti-angiogenic effect of MIG6 in retinal development and neovascularization and the underlying molecular and cellular mechanisms. Loss of function assays using genetic deletion of Mig6 or siRNA knockdown increased angiogenesis in vivo and in vitro, while MIG6 overexpression suppressed pathological angiogenesis. Moreover, we identified the cellular target of MIG6 by revealing its direct inhibitory effect on vascular endothelial cells (ECs). Mechanistically, we found that the anti-angiogenic effect of MIG6 is fulfilled by binding to SHC1 and inhibiting its phosphorylation. Indeed, SHC1 knockdown markedly diminished the effect of MIG6 on ECs. Thus, our findings show that MIG6 is a potent endogenous inhibitor of angiogenesis that may have therapeutic value in anti-angiogenic therapy.
Collapse
Affiliation(s)
- Lixian Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Liying Xing
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Rongyuan Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jianing Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yuye Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lijuan Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Bingbing Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiangrong Ren
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shasha Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Haiqing Kuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xianchai Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Anil Kumar
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jong Kyong Kim
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chunsik Lee
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Venniyoor A. Synergism between anti-angiogenic and immune checkpoint inhibitor drugs: A hypothesis. Med Hypotheses 2020; 146:110399. [PMID: 33239232 DOI: 10.1016/j.mehy.2020.110399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/19/2020] [Accepted: 11/12/2020] [Indexed: 02/08/2023]
Abstract
Hepatocellular cancer (HCC) and renal cell cancer (RCC) are singularly resistant to conventional chemotherapy drugs but therapies targeting the supporting stroma have significantly altered their management. Two recent trials combining anti-angiogenic (AA) agents with immune checkpoint inhibitors (ICIs)- the IMbrave150 and IMmotion151 - have reported impressive progress over targeted agents. It has been suggested that bevacizumab, by improving tissue perfusion, changes the immune suppressive tumour microenvironment to an immune stimulatory one where the ICIs can be more effective. This hypothesis proposes an alternative explanation: That bevacizumab, by increasing tissue hypoxia, amplifies the mutational burden of the tumour by stress-induced mutagenesis, creating a hypermutator profile, which is more vulnerable to the ICI drug, atezolizumab. Additionally, ICIs are known to cause hyperprogression in some tumours, and bevacizumab could provide further benefit by starving these rapidly proliferative tumours of blood supply and nutrients.
Collapse
Affiliation(s)
- Ajit Venniyoor
- National Oncology Centre, The Royal Hospital, Muscat, Oman.
| |
Collapse
|
24
|
Wang M, Du Q, Zuo L, Xue P, Lan C, Sun Z. Metabolism and Distribution of Novel Tumor Targeting Drugs In Vivo. Curr Drug Metab 2020; 21:996-1008. [PMID: 33183197 DOI: 10.2174/1389200221666201112110638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/30/2020] [Accepted: 09/22/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND As a new tumor therapy, targeted therapy is becoming a hot topic due to its high efficiency and low toxicity. Drug effects of targeted tumor drugs are closely related to pharmacokinetics, so it is important to understand their distribution and metabolism in vivo. METHODS A systematic review of the literature on the metabolism and distribution of targeted drugs over the past 20 years was conducted, and the pharmacokinetic parameters of approved targeted drugs were summarized in combination with the FDA's drug instructions. Targeting drugs are divided into two categories: small molecule inhibitors and monoclonal antibodies. Novel targeting drugs and their mechanisms of action, which have been developed in recent years, are summarized. The distribution and metabolic processes of each drug in the human body are reviewed. RESULTS In this review, we found that the distribution and metabolism of small molecule kinase inhibitors (TKI) and monoclonal antibodies (mAb) showed different characteristics based on the differences of action mechanism and molecular characteristics. TKI absorbed rapidly (Tmax ≈ 1-4 h) and distributed in large amounts (Vd > 100 L). It was mainly oxidized and reduced by cytochrome P450 CYP3A4. However, due to the large molecular diameter, mAb was distributed to tissues slowly, and the volume of distribution was usually very low (Vd < 10 L). It was mainly hydrolyzed and metabolized into peptides and amino acids by protease hydrolysis. In addition, some of the latest drugs are still in clinical trials, and the in vivo process still needs further study. CONCLUSION According to the summary of the research progress of the existing targeting drugs, it is found that they have high specificity, but there are still deficiencies in drug resistance and safety. Therefore, the development of safer and more effective targeted drugs is the future research direction. Meanwhile, this study also provides a theoretical basis for clinical accurate drug delivery.
Collapse
Affiliation(s)
- Mengli Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qiuzheng Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lihua Zuo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Xue
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chao Lan
- Department of Emergency Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhi Sun
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
25
|
Jin MZ, Jin WL. The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct Target Ther 2020; 5:166. [PMID: 32843638 PMCID: PMC7447642 DOI: 10.1038/s41392-020-00280-x] [Citation(s) in RCA: 751] [Impact Index Per Article: 150.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/16/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence shows that cellular and acellular components in tumor microenvironment (TME) can reprogram tumor initiation, growth, invasion, metastasis, and response to therapies. Cancer research and treatment have switched from a cancer-centric model to a TME-centric one, considering the increasing significance of TME in cancer biology. Nonetheless, the clinical efficacy of therapeutic strategies targeting TME, especially the specific cells or pathways of TME, remains unsatisfactory. Classifying the chemopathological characteristics of TME and crosstalk among one another can greatly benefit further studies exploring effective treating methods. Herein, we present an updated image of TME with emphasis on hypoxic niche, immune microenvironment, metabolism microenvironment, acidic niche, innervated niche, and mechanical microenvironment. We then summarize conventional drugs including aspirin, celecoxib, β-adrenergic antagonist, metformin, and statin in new antitumor application. These drugs are considered as viable candidates for combination therapy due to their antitumor activity and extensive use in clinical practice. We also provide our outlook on directions and potential applications of TME theory. This review depicts a comprehensive and vivid landscape of TME from biology to treatment.
Collapse
Affiliation(s)
- Ming-Zhu Jin
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China.,Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China.
| |
Collapse
|
26
|
Dai F, Luo F, Zhou R, Zhou Q, Xu J, Zhang Z, Xiao J, Song L. Calponin 3 is associated with poor prognosis and regulates proliferation and metastasis in osteosarcoma. Aging (Albany NY) 2020; 12:14037-14049. [PMID: 32667904 PMCID: PMC7425500 DOI: 10.18632/aging.103224] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 03/31/2020] [Indexed: 04/11/2023]
Abstract
Osteosarcoma is a malignant, life-threatening tumor that affects children and adolescents. In this study, we identified high levels of calponin 3 (CNN3) protein in osteosarcoma tissues and cell lines. The receiver operating characteristic curve analysis revealed that CNN3 has diagnostic value for patients with osteosarcoma. We also found that high CNN3 expression was associated with tumor size, tumor stage, and lymph node and distant metastases. Moreover, high levels of CNN3 mRNA were associated with a poor overall survival rate and a shorter disease-free survival period. CNN3 silencing inhibited cell proliferation, induced apoptosis and cell cycle arrest at the G1 stage, and inhibited cell migration and invasion in vitro. Furthermore, CNN3 silencing also inhibited subcutaneous tumor growth and lung metastasis in vivo. Western blotting revealed that silencing of CNN3 resulted in downregulated expression of MMP9, VEGF, and vimentin, and upregulation of E-cadherin. CNN3 silencing also resulted in downregulation of the ERK1/2 and p38 signaling pathways. In conclusion, high CNN3 expression was found to help in the diagnosis of osteosarcoma, and was found to be associated with poor prognosis in patients. Therefore, CNN3 may play an oncogenic role during the progression of osteosarcoma by activating the ERK1/2 and p38 pathways.
Collapse
Affiliation(s)
- Fei Dai
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Fei Luo
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Rui Zhou
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Qiang Zhou
- Department of Orthopaedics, Third Affliated Hospital, Medical University of Chongqing, Chongqing 401120, China
| | - Jianzhong Xu
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Zehua Zhang
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Jun Xiao
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Lei Song
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| |
Collapse
|
27
|
de Almeida PE, Mak J, Hernandez G, Jesudason R, Herault A, Javinal V, Borneo J, Kim JM, Walsh KB. Anti-VEGF Treatment Enhances CD8 + T-cell Antitumor Activity by Amplifying Hypoxia. Cancer Immunol Res 2020; 8:806-818. [PMID: 32238381 DOI: 10.1158/2326-6066.cir-19-0360] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 11/27/2019] [Accepted: 03/27/2020] [Indexed: 11/16/2022]
Abstract
Antiangiogenic therapies that target the VEGF pathway have been used clinically to combat cancer for over a decade. Beyond having a direct impact on blood vessel development and tumor perfusion, accumulating evidence indicates that these agents also affect antitumor immune responses. Numerous clinical trials combining antiangiogenic drugs with immunotherapies for the treatment of cancer are ongoing, but a mechanistic understanding of how disruption of tumor angiogenesis may impact immunity is not fully discerned. Here, we reveal that blockade of VEGF-A with a mAb to VEGF augments activation of CD8+ T cells within tumors and potentiates their capacity to produce cytokines. We demonstrate that this phenomenon relies on the disruption of VEGFR2 signaling in the tumor microenvironment but does not affect CD8+ T cells directly. Instead, the augmented functional capacity of CD8+ T cells stems from increased tumor hypoxia that initiates a hypoxia-inducible factor-1α program within CD8+ T cells that directly enhances cytokine production. Finally, combinatorial administration of anti-VEGF with an immunotherapeutic antibody, anti-OX40, improved antitumor activity over single-agent treatments. Our findings illustrate that anti-VEGF treatment enhances CD8+ T-cell effector function and provides a mechanistic rationale for combining antiangiogenic and immunotherapeutic drugs for cancer treatment.
Collapse
Affiliation(s)
| | - Judy Mak
- Department of Molecular Oncology, Genentech, Inc., South San Francisco, California
| | - Genevive Hernandez
- Oncology Biomarker Development, Genentech, Inc., South San Francisco, California
| | - Rajiv Jesudason
- Department of Molecular Oncology, Genentech, Inc., South San Francisco, California
| | - Aurelie Herault
- Department of Molecular Oncology, Genentech, Inc., South San Francisco, California
| | - Vincent Javinal
- Department of In-vivo Pharmacology, Genentech, Inc., South San Francisco, California
| | - Jovencio Borneo
- Department of Immunology and Infectious Diseases, Genentech, Inc., South San Francisco, California
| | - Jeong M Kim
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, California
| | - Kevin B Walsh
- Department of Molecular Oncology, Genentech, Inc., South San Francisco, California.
| |
Collapse
|
28
|
Multiple Targets Directed Multiple Ligands: An In Silico and In Vitro Approach to Evaluating the Effect of Triphala on Angiogenesis. Biomolecules 2020; 10:biom10020177. [PMID: 31979409 PMCID: PMC7072423 DOI: 10.3390/biom10020177] [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: 11/06/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis is critical in both physiological and pathological conditions and targeting angiogenesis is a promising strategy for the development of therapies against cancer; however, cells develop resistance to anti-angiogenic therapy, necessitating a more effective strategy. Natural medicines have been used in anti-cancer therapy for many years, but the mechanisms behind these have not generally been explored. Triphala churna (THL), an Indian ayurvedic herbal formulation made from the dried fruits of three medicinal plants, is used as a herbal drug for the treatment of various diseases, including cancer. THL contains over fifteen phytochemicals with different pharmacological effects, especially inhibition of tumor progression. In this study, we examined the effect of these compounds against different targets using docking and in vitro studies. Results showed that THL has a prediction efficacy of (−)436.7, and it inhibited angiogenesis by blocking multiple components of the VEGF/VEGFR2 signaling pathway. The anti-angiogenic effect was mediated by the combined effect of the two top ranked phytochemicals, punicalagin (−424.8) and chebulagic acid (−414.8). The new approach developed in this study to determine the potential efficacy of herbal formulation could be a useful strategy to assess the efficacy of different herbal formulations.
Collapse
|
29
|
Lee HP, Wang SW, Wu YC, Lin LW, Tsai FJ, Yang JS, Li TM, Tang CH. Soya-cerebroside inhibits VEGF-facilitated angiogenesis in endothelial progenitor cells. FOOD AGR IMMUNOL 2020. [DOI: 10.1080/09540105.2020.1713055] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Hsiang-Ping Lee
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yang-Chang Wu
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Liang-Wei Lin
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- China Medical University Children’s Hospital, China Medical University, Taichung, Taiwan
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Te-Mao Li
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| |
Collapse
|
30
|
Mukherjee A, Madamsetty VS, Paul MK, Mukherjee S. Recent Advancements of Nanomedicine towards Antiangiogenic Therapy in Cancer. Int J Mol Sci 2020; 21:E455. [PMID: 31936832 PMCID: PMC7013812 DOI: 10.3390/ijms21020455] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis is a process of generation of de-novo blood vessels from already existing vasculature. It has a crucial role in different physiological process including wound healing, embryonic development, and tumor growth. The methods by which therapeutic drugs inhibit tumor angiogenesis are termed as anti-angiogenesis cancer therapy. Developments of angiogenic inhibiting drugs have various limitations causing a barrier for successful treatment of cancer, where angiogenesis plays an important role. In this context, investigators developed novel strategies using nanotechnological approaches that have demonstrated inherent antiangiogenic properties or used for the delivery of antiangiogenic agents in a targeted manner. In this present article, we decisively highlight the recent developments of various nanoparticles (NPs) including liposomes, lipid NPs, protein NPs, polymer NPs, inorganic NPs, viral and bio-inspired NPs for potential application in antiangiogenic cancer therapy. Additionally, the clinical perspectives, challenges of nanomedicine, and future perspectives are briefly analyzed.
Collapse
Affiliation(s)
- Anubhab Mukherjee
- Aavishkar Oral Strips Pvt Ltd., 109/3, IDA, Phase 2, Sector 2, Lane 6, Cherlapally, Hyderabad 500051, India;
| | - Vijay Sagar Madamsetty
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA;
| | - Manash K. Paul
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, The University of California, Los Angeles (UCLA), Factor Bldg. 10-240, 621 Charles E. Young Dr., Los Angeles, CA 90095, USA
| | - Sudip Mukherjee
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| |
Collapse
|
31
|
Zamorano P, Koning T, Oyanadel C, Mardones GA, Ehrenfeld P, Boric MP, González A, Soza A, Sánchez FA. Galectin-8 induces endothelial hyperpermeability through the eNOS pathway involving S-nitrosylation-mediated adherens junction disassembly. Carcinogenesis 2019; 40:313-323. [PMID: 30624618 DOI: 10.1093/carcin/bgz002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 12/04/2018] [Accepted: 01/04/2019] [Indexed: 12/26/2022] Open
Abstract
The permeability of endothelial cells is regulated by the stability of the adherens junctions, which is highly sensitive to kinase-mediated phosphorylation and endothelial nitric oxide synthase (eNOS)-mediated S-nitrosylation of its protein components. Solid tumors can produce a variety of factors that stimulate these signaling pathways leading to endothelial cell hyperpermeability. This generates stromal conditions that facilitate tumoral growth and dissemination. Galectin-8 (Gal-8) is overexpressed in several carcinomas and has a variety of cellular effects that can contribute to tumor pathogenicity, including angiogenesis. Here we explored whether Gal-8 has also a role in endothelial permeability. We show that recombinant Gal-8 activates eNOS, induces S-nitrosylation of p120-catenin (p120) and dissociation of adherens junction, leading to hyperpermeability of the human endothelial cell line EAhy926. This pathway involves focal-adhesion kinase (FAK) activation downstream of eNOS as a requirement for eNOS-mediated p120 S-nitrosylation. This suggests a reciprocal, yet little understood, regulation of phosphorylation and S-nitrosylation events acting upon adherens junction permeability. In addition, glutathione S-transferase (GST)-Gal-8 pull-down experiments and function-blocking β1-integrin antibodies point to β1-integrins as cell surface components involved in Gal-8-induced hyperpermeability. Endogenous Gal-8 secreted from the breast cancer cell line MCF-7 has similar hyperpermeability and signaling effects. Furthermore, the mouse cremaster model system showed that Gal-8 also activates eNOS, induces S-nitrosylation of adherens junction components and is an effective hyperpermeability agent in vivo. These results add endothelial permeability regulation by S-nitrosylation as a new function of Gal-8 that can potentially contribute to the pathogenicity of tumors overexpressing this lectin.
Collapse
Affiliation(s)
- Patricia Zamorano
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Tania Koning
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Claudia Oyanadel
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Gonzalo A Mardones
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Instituto de Fisiología, Valdivia, Chile.,Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Valdivia, Chile
| | - Pamela Ehrenfeld
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Valdivia, Chile.,Instituto de Anatomía, Histología y Patología, Universidad Austral de Chile, Valdivia, Chile
| | | | - Alfonso González
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Centro de Envejecimiento y Regeneración, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Fundación Ciencia y Vida. Santiago, Chile
| | - Andrea Soza
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Centro de Envejecimiento y Regeneración, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fabiola A Sánchez
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia 5110566, Chile.,Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Valdivia, Chile
| |
Collapse
|
32
|
Wang L, Wei CY, Xu YY, Deng XY, Wang Q, Ying JH, Zhang SM, Yuan X, Xuan TF, Pan YY, Gu JY. Prognostic genes of melanoma identified by weighted gene co-expression network analysis and drug repositioning using a network-based method. Oncol Lett 2019; 18:6066-6078. [PMID: 31788081 PMCID: PMC6864934 DOI: 10.3892/ol.2019.10961] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 08/21/2019] [Indexed: 12/19/2022] Open
Abstract
Melanoma is one of the most malignant types of skin cancer. However, the efficacy and utility of available drug therapies for melanoma are limited. The objective of the present study was to identify potential genes associated with melanoma progression and to explore approved therapeutic drugs that target these genes. Weighted gene co-expression network analysis was used to construct a gene co-expression network, explore the associations between genes and clinical characteristics and identify potential biomarkers. Gene expression profiles of the GSE65904 dataset were obtained from the Gene Expression Omnibus database. RNA-sequencing data and clinical information associated with melanoma obtained from The Cancer Genome Atlas were used for biomarker validation. A total of 15 modules were identified through average linkage hierarchical clustering. In the two significant modules, three network hub genes associated with melanoma prognosis were identified: C-X-C motif chemokine receptor 4 (CXCR4), interleukin 7 receptor (IL7R) and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit γ (PIK3CG). The receiver operating characteristic curve indicated that the mRNA levels of these genes exhibited excellent prognostic efficiency for primary and metastatic tumor tissues. In addition, the proximity between candidate genes associated with melanoma progression and drug targets obtained from DrugBank was calculated in the protein interaction network, and the top 15 drugs that may be suitable for treating melanoma were identified. In summary, co-expression network analysis led to the selection of CXCR4, IL7R and PIK3CG for further basic and clinical research on melanoma. Utilizing a network-based method, 15 drugs that exhibited potential for the treatment of melanoma were identified.
Collapse
Affiliation(s)
- Lu Wang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Chuan-Yuan Wei
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yuan-Yuan Xu
- Department of Surgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xin-Yi Deng
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Qiang Wang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Jiang-Hui Ying
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Si-Min Zhang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Xin Yuan
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Tian-Fan Xuan
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yu-Yan Pan
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Jian-Ying Gu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
33
|
Pezzella F. Mechanisms of resistance to anti-angiogenic treatments. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:595-607. [PMID: 35582580 PMCID: PMC8992538 DOI: 10.20517/cdr.2019.39] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 06/25/2019] [Accepted: 07/02/2019] [Indexed: 05/31/2023]
Abstract
Hailed as the cancer treatment to end all the resistance to treatment, anti-angiogenic therapy turned out to be not quite what was promised. The hope that this therapeutic approach would not have suffered by the phenomenon of resistance was based on the fact that was targeting normal vessels rather than tumour cells prone to mutation and subject to drug induced selection. However, reality turned out to be more complex and since 1997, several mechanisms of resistance have been described to the point that the study of resistance to these drugs is now a very large field. Far from being exhaustive, this paper presents the main mechanisms discovered trough some examples.
Collapse
Affiliation(s)
- Francesco Pezzella
- Nuffield Division of Clinical Laboratory Science, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| |
Collapse
|
34
|
Inactivation of Rab27B-dependent signaling pathway by calycosin inhibits migration and invasion of ER-negative breast cancer cells. Gene 2019; 709:48-55. [PMID: 31002890 DOI: 10.1016/j.gene.2019.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 03/25/2019] [Accepted: 04/02/2019] [Indexed: 01/08/2023]
|
35
|
Ali Z, Mukwaya A, Biesemeier A, Ntzouni M, Ramsköld D, Giatrellis S, Mammadzada P, Cao R, Lennikov A, Marass M, Gerri C, Hildesjö C, Taylor M, Deng Q, Peebo B, del Peso L, Kvanta A, Sandberg R, Schraermeyer U, Andre H, Steffensen JF, Lagali N, Cao Y, Kele J, Jensen LD. Intussusceptive Vascular Remodeling Precedes Pathological Neovascularization. Arterioscler Thromb Vasc Biol 2019; 39:1402-1418. [PMID: 31242036 PMCID: PMC6636809 DOI: 10.1161/atvbaha.118.312190] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Supplemental Digital Content is available in the text. Objective— Pathological neovascularization is crucial for progression and morbidity of serious diseases such as cancer, diabetic retinopathy, and age-related macular degeneration. While mechanisms of ongoing pathological neovascularization have been extensively studied, the initiating pathological vascular remodeling (PVR) events, which precede neovascularization remains poorly understood. Here, we identify novel molecular and cellular mechanisms of preneovascular PVR, by using the adult choriocapillaris as a model. Approach and Results— Using hypoxia or forced overexpression of VEGF (vascular endothelial growth factor) in the subretinal space to induce PVR in zebrafish and rats respectively, and by analyzing choriocapillaris membranes adjacent to choroidal neovascular lesions from age-related macular degeneration patients, we show that the choriocapillaris undergo robust induction of vascular intussusception and permeability at preneovascular stages of PVR. This PVR response included endothelial cell proliferation, formation of endothelial luminal processes, extensive vesiculation and thickening of the endothelium, degradation of collagen fibers, and splitting of existing extravascular columns. RNA-sequencing established a role for endothelial tight junction disruption, cytoskeletal remodeling, vesicle- and cilium biogenesis in this process. Mechanistically, using genetic gain- and loss-of-function zebrafish models and analysis of primary human choriocapillaris endothelial cells, we determined that HIF (hypoxia-induced factor)-1α-VEGF-A-VEGFR2 signaling was important for hypoxia-induced PVR. Conclusions— Our findings reveal that PVR involving intussusception and splitting of extravascular columns, endothelial proliferation, vesiculation, fenestration, and thickening is induced before neovascularization, suggesting that identifying and targeting these processes may prevent development of advanced neovascular disease in the future.
Collapse
Affiliation(s)
- Zaheer Ali
- From the Division of Cardiovascular Medicine, Department of Medical and Health Sciences (Z.A., L.D.J.), Linkoping University, Sweden
| | - Anthony Mukwaya
- Division of Ophthalmology, Department of Clinical and Experimental Medicine (A.M., A.L., B.P., N.L.), Linkoping University, Sweden
| | - Antje Biesemeier
- Experimental Vitreoretinal Surgery, Center for Ophthalmology, University of Tuebingen, Germany (A.B., U.S.)
| | - Maria Ntzouni
- Electronmicroscopy and Histology Laboratory, Faculty of Medicine (M.N.), Linkoping University, Sweden
| | - Daniel Ramsköld
- Department of Cell and Molecular Biology (D.R., S.G., R.S.), Karolinska Institutet, Stockholm, Sweden
| | - Sarantis Giatrellis
- Department of Cell and Molecular Biology (D.R., S.G., R.S.), Karolinska Institutet, Stockholm, Sweden
| | - Parviz Mammadzada
- Department of Clinical Neuroscience, Section for Ophthalmology and Vision, St. Erik Eye Hospital (P.M., A.K., H.A.), Karolinska Institutet, Stockholm, Sweden
| | - Renhai Cao
- Department of Microbiology, Tumor and Cell Biology (R.C., Y.C.), Karolinska Institutet, Stockholm, Sweden
| | - Anton Lennikov
- Division of Ophthalmology, Department of Clinical and Experimental Medicine (A.M., A.L., B.P., N.L.), Linkoping University, Sweden
| | - Michele Marass
- Department of Developmental Genetics, Max Planck Institute for Lung and Heart Research, Bad Nauheim, Germany (M.M., C.G.)
| | - Claudia Gerri
- Department of Developmental Genetics, Max Planck Institute for Lung and Heart Research, Bad Nauheim, Germany (M.M., C.G.)
| | - Camilla Hildesjö
- Division of Surgery, Orthopedics and Oncology, Department for Clinical and Experimental Medicine (C.H.), Linkoping University, Sweden
| | - Michael Taylor
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison (M.T.)
| | - Qiaolin Deng
- Department of Physiology and Pharmacology (Q.D., J.K.), Karolinska Institutet, Stockholm, Sweden
| | - Beatrice Peebo
- Division of Ophthalmology, Department of Clinical and Experimental Medicine (A.M., A.L., B.P., N.L.), Linkoping University, Sweden
| | - Luis del Peso
- Department of Biochemistry, Universidad Autónoma de Madrid, Spain (L.d.P.)
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM Madrid, Spain (L.d.P.)
| | - Anders Kvanta
- Department of Clinical Neuroscience, Section for Ophthalmology and Vision, St. Erik Eye Hospital (P.M., A.K., H.A.), Karolinska Institutet, Stockholm, Sweden
| | - Rickard Sandberg
- Department of Cell and Molecular Biology (D.R., S.G., R.S.), Karolinska Institutet, Stockholm, Sweden
| | - Ulrich Schraermeyer
- Experimental Vitreoretinal Surgery, Center for Ophthalmology, University of Tuebingen, Germany (A.B., U.S.)
| | - Helder Andre
- Department of Clinical Neuroscience, Section for Ophthalmology and Vision, St. Erik Eye Hospital (P.M., A.K., H.A.), Karolinska Institutet, Stockholm, Sweden
| | - John F. Steffensen
- Marine Biological Section, Biological Institute, University of Copenhagen, Helsingor, Denmark (J.F.S.)
| | - Neil Lagali
- Division of Ophthalmology, Department of Clinical and Experimental Medicine (A.M., A.L., B.P., N.L.), Linkoping University, Sweden
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology (R.C., Y.C.), Karolinska Institutet, Stockholm, Sweden
| | - Julianna Kele
- Department of Physiology and Pharmacology (Q.D., J.K.), Karolinska Institutet, Stockholm, Sweden
| | - Lasse Dahl Jensen
- From the Division of Cardiovascular Medicine, Department of Medical and Health Sciences (Z.A., L.D.J.), Linkoping University, Sweden
| |
Collapse
|
36
|
Li Y, Wan YY, Zhu B. Immune Cell Metabolism in Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1011:163-196. [PMID: 28875490 DOI: 10.1007/978-94-024-1170-6_5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tumor microenvironment (TME) is composed of tumor cells, immune cells, cytokines, extracellular matrix, etc. The immune system and the metabolisms of glucose, lipids, amino acids, and nucleotides are integrated in the tumorigenesis and development. Cancer cells and immune cells show metabolic reprogramming in the TME, which intimately links immune cell functions and edits tumor immunology. Recent findings in immune cell metabolism hold the promising possibilities toward clinical therapeutics for treating cancer. This chapter introduces the updated understandings of metabolic reprogramming of immune cells in the TME and suggests new directions in manipulation of immune responses for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Yongsheng Li
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yisong Y Wan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
37
|
Amerizadeh F, Khazaei M, Maftouh M, Mardani R, Bahrami A. miRNA Targeting Angiogenesis as a Potential Therapeutic Approach in the Treatment of Colorectal Cancers. Curr Pharm Des 2019; 24:4668-4674. [DOI: 10.2174/1381612825666190110161843] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/27/2018] [Accepted: 01/01/2019] [Indexed: 12/11/2022]
Abstract
Angiogenesis refers to the formation of recent blood vessels, which is one of the characteristics of
cancer progression and it has been deliberated as a putative target to the treatment of many kinds of cancers. The
VEGF signaling substrate is very important for angiogenesis and is commonly high-regulated in tumors. As a
result, this molecule has attracted the attention of most of the researchers to develop antiangiogenic therapies. We
have presented that VEGF blockage in neoadjuvant setting via bevacizumab, aflibercept and sunitinib not only
has revealed some promising benefits but also has shown a large negative outcome in the adjuvant trials. However,
at an advanced stage of tumors, suppression of VEGF alone is inadequate to stop advancement, encouraging
drug resistance, and probably enhancing metastasis and invasion in the tumor microenvironment, thereby suggesting
the therapeutic potential of targeting angiogenic pathways in gastrointestinal cancers.
Collapse
Affiliation(s)
- Forouzan Amerizadeh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Maftouh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramin Mardani
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Afsane Bahrami
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
38
|
Chiang KC, Huang ST, Wu RC, Huang SC, Yeh TS, Chen MH, Hsu JT, Chen LW, Kuo SF, Chueh HY, Juang HH, Hung SI, Yeh CN, Pang JHS. Interferon α-inducible protein 27 is an oncogene and highly expressed in cholangiocarcinoma patients with poor survival. Cancer Manag Res 2019; 11:1893-1905. [PMID: 30881116 PMCID: PMC6400119 DOI: 10.2147/cmar.s196485] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE Cholangiocarcinoma (CCA) is a devastating disease. Interferon α-inducible protein 27 (IFI27), originally known to involve in innate immunity, is later found to intervene in cell proliferation, leading to inventive studies regarding the role of IFI27 in cancer treatment. We aimed to investigate the role of IFI27 in CCA. MATERIALS AND METHODS Cell proliferation, migration, and invasion assays, Western blot, gene transfection and knockdown, immunofluorescent and immunohistochemical stains, and xenograft animal model were applied. RESULTS IFI27 knockdown in CCA cells induced cell cycle arrest in S phase, resulting in lower cell proliferative rate in vitro and in vivo. IFI27 knockdown attenuated CCA cell migration and invasion through inhibition of epithelial-mesenchymal transition, which was supported by increased E-cadherin and decreased N-cadherin and fibronectin. Filamentous actin level was also reduced. IFI27 knockdown further repressed expression and secretion of vascular endothelial growth factor (VEGF-A), a strong stimulator of angiogenesis, through downregulation of c-jun and c-fos, which was supported in vitro by the finding that human vascular endothelial cells grew more slowly in conditioned medium of IFI27 knockdown on CCA cells and in vivo by the lower erythropoietin concentration found in the xenografted tumors derived from IFI27 knockdown on CCA cells. In addition, anti-VEGF-A antibody treatment was able to repress CCA cell growth. To the contrary, IFI27 overexpression could increase CCA cell proliferation, migration, and invasion. Clinically, higher IFI27 expression was linked to inferior overall survival of CCA patients. CONCLUSION Our data strongly suggest that IFI27 could be deemed as a potential target for CCA treatment.
Collapse
Affiliation(s)
- Kun-Chun Chiang
- General Surgery Department, Chang Gung, Memorial Hospital, Chang Gung University, Keelung, Taiwan, ROC
| | - Sheng-Teng Huang
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan, ROC
| | - Ren-Chin Wu
- Department of Anatomic Pathology, Chang Gung Memorial Hospital, Kwei-Shan, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Shih-Chiang Huang
- Department of Anatomic Pathology, Chang Gung Memorial Hospital, Kwei-Shan, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Ta-Sen Yeh
- General Surgery Department, Chang Gung Memorial Hospital, Kwei-Shan, Chang Gung University, Taoyuan, Taiwan, ROC,
| | - Ming-Huang Chen
- Division of Hematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Jun-Te Hsu
- General Surgery Department, Chang Gung Memorial Hospital, Kwei-Shan, Chang Gung University, Taoyuan, Taiwan, ROC,
| | - Li-Wei Chen
- Department of Gastroenterology, Chang Gung Memorial Hospital, Chang Gung University, Keelung, Taiwan, ROC
| | - Sheng-Fong Kuo
- Department of Endocrinology and Metabolism, Chang Gung Memorial Hospital, Chang Gung University, Keelung, Taiwan, ROC
| | - Ho-Yen Chueh
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| | - Horng-Heng Juang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan, ROC
| | - Shuen-Iu Hung
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Chun-Nan Yeh
- General Surgery Department, Chang Gung Memorial Hospital, Kwei-Shan, Chang Gung University, Taoyuan, Taiwan, ROC,
| | - Jong-Hwei S Pang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan, Taiwan, ROC,
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Linkow, Taoyuan City, Taiwan, ROC,
| |
Collapse
|
39
|
Moehler T, Hose D, Andrulis M, Seckinger A, Goldschmidt H. The Value of Anti-angiogenics in Multiple Myeloma Therapy. TUMOR ANGIOGENESIS 2019:639-658. [DOI: 10.1007/978-3-319-33673-2_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
40
|
ZHOU Y, CAO HB, LI WJ, ZHAO L. The CXCL12 (SDF-1)/CXCR4 chemokine axis: Oncogenic properties, molecular targeting, and synthetic and natural product CXCR4 inhibitors for cancer therapy. Chin J Nat Med 2018; 16:801-810. [DOI: 10.1016/s1875-5364(18)30122-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Indexed: 02/07/2023]
|
41
|
Tomida C, Yamagishi N, Nagano H, Uchida T, Ohno A, Hirasaka K, Nikawa T, Teshima-Kondo S. Antiangiogenic agent sunitinib induces epithelial to mesenchymal transition and accelerates motility of colorectal cancer cells. THE JOURNAL OF MEDICAL INVESTIGATION 2018; 64:250-254. [PMID: 28954991 DOI: 10.2152/jmi.64.250] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Although vascular endothelial growth factor receptor (VEGF-R)-targeted antiangiogenic agents are important treatment for a number of human malignancies, there is accumulating evidence that the therapies may promote disease progression, such as invasion and metastasis. How tumors become to promote their evasiveness remains fully uncertain. One of possible mechanisms for the adaptation may be a direct effect of VEGF-R inhibitors on tumor cells expressing VEGF-R. To elucidate a direct effect of VEGF-R-targeting drug (sunitinib), we established a human colorectal cancer cell model adapted to sunitinib. The sunitinib-conditioned cells showed a significant increase in cellular motility and migration activities, compared to the vehicle-treated control cells. Consistent with the phenotype, the sunitinib-conditioned cells decreased the expression levels of E-cadherin (an epithelial marker), while significantly increased the levels of Slug and Zeb1 (mesenchymal markers). Expression profiles of VEGF-R in the sunitinib-conditioned cells showed that only neuropilin-1 (NRP1) expression was significantly increased among all VEGF-R tested. Blockade of NRP1 using its antagonist clearly repressed the migration activation in sunitinib-conditioned cells, but not in the control cells. These results suggest that inhibition of VEGF-R on colorectal cancer cells can drive the epithelial-mesenchymal transition, leading to activation of cell motility in an NRP1-dependent manner. J. Med. Invest. 64: 250-254, August, 2017.
Collapse
Affiliation(s)
- Chisato Tomida
- Department of Physiological Nutrition, Institute of Medical Nutrition, University of Tokushima Graduate School
| | - Naoko Yamagishi
- Department of Anatomy and Cell Biology, School of Medicine, Wakayama Medical University
| | - Hikaru Nagano
- Department of Physiological Nutrition, Institute of Medical Nutrition, University of Tokushima Graduate School
| | - Takayuki Uchida
- Department of Physiological Nutrition, Institute of Medical Nutrition, University of Tokushima Graduate School
| | - Ayako Ohno
- Department of Physiological Nutrition, Institute of Medical Nutrition, University of Tokushima Graduate School
| | - Katsuya Hirasaka
- Graduate school of Fisheries Science and Environmental Studies, Nagasaki University
| | - Takeshi Nikawa
- Department of Physiological Nutrition, Institute of Medical Nutrition, University of Tokushima Graduate School
| | - Shigetada Teshima-Kondo
- Department of Physiological Nutrition, Institute of Medical Nutrition, University of Tokushima Graduate School
| |
Collapse
|
42
|
El Alaoui-Lasmaili K, Faivre B. Antiangiogenic therapy: Markers of response, "normalization" and resistance. Crit Rev Oncol Hematol 2018; 128:118-129. [PMID: 29958627 DOI: 10.1016/j.critrevonc.2018.06.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 02/08/2023] Open
Abstract
Currently in cancer treatment, one premise is to use antiangiogenic therapies in association with chemotherapy or radiotherapy to augment their efficacy by benefiting from the vascular "normalization" induced by antiangiogenic therapy. This concept defines the time during which the tumor blood vessels adopt normal-like morphology and functionality, i.e. the blood vessels become more mature, the perfusion augments and hypoxia decreases. To date, there is such a diversity of treatment protocols where the type of antiangiogenic to adopt, its dose and duration of administration are different, that knowing when and how to treat is problematic. In this review, we analyzed thoroughly preclinical and clinical studies that use antiangiogenic treatments to benefit from the "normalization" and showed that the effects depend on the type of antiangiogenic administrated (anti-VEGF, anti-VEGFR, Multi-Kinase Inhibitor) and on the duration of treatment. Finally, biomarkers of "normalization" and resistance that could be used in the clinic are presented.
Collapse
Affiliation(s)
| | - Béatrice Faivre
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; Université de Lorraine, Faculté de Pharmacie, Nancy, France.
| |
Collapse
|
43
|
Rhodocetin-αβ selectively breaks the endothelial barrier of the tumor vasculature in HT1080 fibrosarcoma and A431 epidermoid carcinoma tumor models. Oncotarget 2018; 9:22406-22422. [PMID: 29854288 PMCID: PMC5976474 DOI: 10.18632/oncotarget.25032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 03/19/2018] [Indexed: 11/29/2022] Open
Abstract
The tumor vasculature differs from normal blood vessels in morphology, composition and stability. Here, we describe a novel tumor vessel-disrupting mechanism. In an HT1080/mouse xenograft tumor model rhodocetin-αβ was highly effective in disrupting the tumor endothelial barrier. Mechanistically, rhodocetin-αβ triggered MET signaling via neuropilin-1. As both neuropilin-1 and MET were only lumen-exposed in a subset of abnormal tumor vessels, but not in normal vessels, the prime target of rhodocetin-αβ were these abnormal tumor vessels. Consequently, cells lining such tumor vessels became increasingly motile which compromised the vessel wall tightness. After this initial leakage, rhodocetin-αβ could leave the bloodstream and reach the as yet inaccessible neuropilin-1 on the basolateral side of endothelial cells and thus disrupt nearby vessels. Due to the specific neuropilin-1/MET co-distribution on cells lining such abnormal tumor vessels in contrast to normal endothelial cells, rhodocetin-αβ formed the necessary trimeric signaling complex of rhodocetin-αβ-MET-neuropilin-1 only in these abnormal tumor vessels. This selective attack of tumor vessels, sparing endothelial cell-lined vessels of normal tissues, suggests that the neuropilin-1-MET signaling axis may be a promising drugable target for anti-tumor therapy, and that rhodocetin-αβ may serve as a lead structure to develop novel anti-tumor drugs that target such vessels.
Collapse
|
44
|
Wang T, Chen S, Wang S, Shi L, Wang C, Zhang J, Gao Y, Li G, Qi Y, An X, Chen L. Targeting neurokinin-3 receptor: a novel anti-angiogenesis strategy for cancer treatment. Oncotarget 2018; 8:40713-40723. [PMID: 28489574 PMCID: PMC5522309 DOI: 10.18632/oncotarget.17250] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 04/06/2017] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis is essential for tumor growth and metastasis, controlling angiogenesis is a promising strategy in cancer treatment. However, thus farther severe side effects of anti-angiogenic drugs have been rather demonstrated, stimulating interest in seeking novel targets of anti-angiogenesis. Neurokinin receptors, also known as tachykinin receptors, are usually considered as drug targets due to diverse physiological functions and their tractability. Although Neurokinin B, the selective natural agonist of neurokinin-3 receptor, have been shown to exhibit anti-angiogenesis activity, the effect and mechanism of neurokinin-3 receptor-mediated angiogenesis still remains unclear. In the present study, we demonstrated that [Mephe7]NKB, an analogue of NKB, possess significant anti-angiogenic effect on CAM. Furthermore, by introducing the tumor angiogenesis homing sequence (NGR), we designed and synthesized two novel agonist analogues of NK3R, NK3R-A1 and NK3R-A2. Both of the two analogues exhibit more efficient anti-migration effect on HUVECs by activating NK3R in vitro, and showed potent antitumor activities with no significant side effects in vivo. Taken together, our results illuminated that NK3R might be a potential novel target for the anti-angiogenesis therapy. Notably, NK3R-A1 might be used as a template for the development of the anti-tumor drugs on the basis of the anti-angiogenesis strategy.
Collapse
Affiliation(s)
- Ting Wang
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P.R. China
| | - Siwei Chen
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P.R. China
| | - Shihui Wang
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P.R. China
| | - Liang Shi
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P.R. China
| | - Chenggong Wang
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P.R. China
| | - Jingxin Zhang
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P.R. China
| | - Yanfeng Gao
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P.R. China
| | - Guodong Li
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P.R. China
| | - Yuanming Qi
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P.R. China
| | - Xiuli An
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P.R. China
| | - Lixiang Chen
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P.R. China
| |
Collapse
|
45
|
Lin CY, Tzeng HE, Li TM, Chen HT, Lee Y, Yang YC, Wang SW, Yang WH, Tang CH. WISP-3 inhibition of miR-452 promotes VEGF-A expression in chondrosarcoma cells and induces endothelial progenitor cells angiogenesis. Oncotarget 2018; 8:39571-39581. [PMID: 28465477 PMCID: PMC5503633 DOI: 10.18632/oncotarget.17142] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/29/2017] [Indexed: 12/24/2022] Open
Abstract
Chondrosarcoma is the second most prevalent general primary tumor of bone following osteosarcoma. Chondrosarcoma development may be linked to angiogenesis, which is principally elicited by vascular endothelial growth factor-A (VEGF-A). VEGF-A level has been recognized as a prognostic marker in angiogenesis. WNT1-inducible signaling pathway protein-3 (WISP)-3/CCN6 belongs to the CCN family and is involved in regulating several cellular functions, including cell proliferation, differentiation, and migration. Nevertheless, the effect of WISP-3 on VEGF-A production and angiogenesis in human chondrosarcoma remains largely unknown. This current study shows that WISP-3 promoted VEGF-A production and induced angiogenesis of human endothelial progenitor cells. Moreover, WISP-3-enhanced VEGF-A expression and angiogenesis involved the c-Src and p38 signaling pathways, while miR-452 expression was negatively affected by WISP-3 via the c-Src and p38 pathways. Our results illustrate the clinical significance of WISP-3, VEGF-A and miR-452 in human chondrosarcoma patients. WISP-3 may illustrate a novel therapeutic target in the metastasis and angiogenesis of chondrosarcoma.
Collapse
Affiliation(s)
- Chih-Yang Lin
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Huey-En Tzeng
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Department of Internal Medicine, Division of Hematology and Oncology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Te-Mao Li
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Hsien-Te Chen
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Yi Lee
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Yi-Chen Yang
- Department of Nursing, National Taichung University of Science and Technology, Taichung, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Wei-Hung Yang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,Department of Nursing, National Taichung University of Science and Technology, Taichung, Taiwan.,Department of Orthopedic Surgery, Taichung Hospital, Ministry of Health and Welfare, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
46
|
Tomida C, Yamagishi N, Nagano H, Uchida T, Ohno A, Hirasaka K, Nikawa T, Teshima-Kondo S. VEGF pathway-targeting drugs induce evasive adaptation by activation of neuropilin-1/cMet in colon cancer cells. Int J Oncol 2018. [PMID: 29532881 DOI: 10.3892/ijo.2018.4291] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Anti-angiogenic therapies targeting vascular endothelial growth factor (VEGF) and its receptor (VEGF-R) are important treatments for a number of human malignancies, including colorectal cancers. However, there is increasing evidence that VEGF/VEGF-R inhibitors promote the adaptive and evasive resistance of tumor cells to the therapies. The mechanism by which the cancer cells become resistant remains unclear. One potential mechanism is that VEGF/VEGF-R blockers directly act on tumor cells independently of anti-angiogenic effects. In this study, the direct effects of an anti-VEGF antibody (bevacizumab) and a VEGF-R tyrosine kinase inhibitor (sunitinib) on the evasive adaptation of colon cancer cells were compared. HCT116 and RKO human colon cancer cell lines were chronically exposed (3 months) to bevacizumab or sunitinib in vitro to establish bevacizumab- and sunitinib-adapted cells, respectively. Transwell migration and invasion assays, western blotting, reverse transcription-quantitative polymerase chain reaction, co-immunoprecipitation analysis, cell survival assays and ELISAs were conducted to analyze the adapted cells. Compared with the control vehicle-treated cells, the two cell models exhibited increased migration and invasion activities to different degrees and through different mechanisms. The bevacizumab-adapted cells, but not in the sunitinib-adapted cells, exhibited redundantly increased expression levels of VEGF/VEGF-R family members, including VEGF-A, placental growth factor, VEGF-C, VEGF-R1 and VEGF-R3. In addition, the phosphorylation levels of VEGF-R1 and VEGF-R3 were increased in the bevacizumab-adapted cells compared with the control cells. Thus, the inhibition of VEGF-R1 and VEGF-R3 decreased the evasive activities of the cells, suggesting that they remained dependent on redundant VEGF/VEGF-R signaling. By contrast, the sunitinib-adapted cells exhibited increased neuropilin-1 (NRP1) expression levels compared with the control cells. In the sunitinib-adapted cells, NRP1 interacted with phosphorylated cMet, and the cMet activation was dependent on NRP1. Thus, NRP1 or cMet blockade suppressed the evasive activation of the sunitinib-adapted cells. These results suggest that the sunitinib-adapted cells switched from a VEGF-R-dependent pathway to an alternative NRP1/cMet-dependent one. The findings of the present study indicate that VEGF/VEGF-R inhibitors directly act on colon cancer cells and activate their evasive adaptation via different mechanisms.
Collapse
Affiliation(s)
- Chisato Tomida
- Department of Physiological Nutrition, Institute of Medical Nutrition, University of Tokushima Graduate School, Kuramoto, Tokushima 770-8503, Japan
| | - Naoko Yamagishi
- Department of Anatomy and Cell Biology, School of Medicine, Wakayama Medical University, Kimiidera, Wakayama 641-8509, Japan
| | - Hikaru Nagano
- Department of Clinical Nutrition, Osaka Prefecture University Graduate School, Habikino, Osaka 583-8555, Japan
| | - Takayuki Uchida
- Department of Physiological Nutrition, Institute of Medical Nutrition, University of Tokushima Graduate School, Kuramoto, Tokushima 770-8503, Japan
| | - Ayako Ohno
- Department of Physiological Nutrition, Institute of Medical Nutrition, University of Tokushima Graduate School, Kuramoto, Tokushima 770-8503, Japan
| | - Katsuya Hirasaka
- Graduate School of Fisheries Science and Environmental Studies, Nagasaki University, Nagasaki, Nagasaki 852-8521, Japan
| | - Takeshi Nikawa
- Department of Physiological Nutrition, Institute of Medical Nutrition, University of Tokushima Graduate School, Kuramoto, Tokushima 770-8503, Japan
| | - Shigetada Teshima-Kondo
- Department of Clinical Nutrition, Osaka Prefecture University Graduate School, Habikino, Osaka 583-8555, Japan
| |
Collapse
|
47
|
Arreola A, Payne LB, Julian MH, de Cubas AA, Daniels AB, Taylor S, Zhao H, Darden J, Bautch VL, Rathmell WK, Chappell JC. Von Hippel-Lindau mutations disrupt vascular patterning and maturation via Notch. JCI Insight 2018; 3:92193. [PMID: 29467323 DOI: 10.1172/jci.insight.92193] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 01/18/2018] [Indexed: 12/27/2022] Open
Abstract
Von Hippel-Lindau (VHL) gene mutations induce neural tissue hemangioblastomas, as well as highly vascularized clear cell renal cell carcinomas (ccRCCs). Pathological vessel remodeling arises from misregulation of HIFs and VEGF, among other genes. Variation in disease penetrance has long been recognized in relation to genotype. We show Vhl mutations also disrupt Notch signaling, causing mutation-specific vascular abnormalities, e.g., type 1 (null) vs. type 2B (murine G518A representing human R167Q). In conditional mutation retina vasculature, Vhl-null mutation (i.e., UBCCreER/+Vhlfl/fl) had little effect on initial vessel branching, but it severely reduced arterial and venous branching at later stages. Interestingly, this mutation accelerated arterial maturation, as observed in retina vessel morphology and aberrant α-smooth muscle actin localization, particularly in vascular pericytes. RNA sequencing analysis identified gene expression changes within several key pathways, including Notch and smooth muscle cell contractility. Notch inhibition failed to reverse later-stage branching defects but rescued the accelerated arterialization. Retinal vessels harboring the type 2B Vhl mutation (i.e., UBCCreER/+Vhlfl/2B) displayed stage-specific changes in vessel branching and an advanced progression toward an arterial phenotype. Disrupting Notch signaling in type 2B mutants increased both artery and vein branching and restored arterial maturation toward nonmutant levels. By revealing differential effects of the null and type 2B Vhl mutations on vessel branching and maturation, these data may provide insight into the variability of VHL-associated vascular changes - particularly the heterogeneity and aggressiveness in ccRCC vessel growth - and also suggest Notch pathway targets for treating VHL syndrome.
Collapse
Affiliation(s)
- Alexandra Arreola
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, North Carolina, USA
| | | | - Morgan H Julian
- Center for Heart and Regenerative Medicine and.,Department of Basic Science Education, Virginia Tech Carilion School of Medicine and Research Institute, Roanoke, Virginia, USA
| | | | - Anthony B Daniels
- Department of Ophthalmology and Visual Sciences.,Department of Biochemistry.,Department of Radiation Oncology, and.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Huaning Zhao
- Center for Heart and Regenerative Medicine and.,Department of Biomedical Engineering and Mechanics
| | - Jordan Darden
- Center for Heart and Regenerative Medicine and.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Victoria L Bautch
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, North Carolina, USA.,Department of Biology and.,McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, USA
| | - W Kimryn Rathmell
- Department of Medicine, Division of Hematology and Oncology.,Department of Biochemistry.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John C Chappell
- Center for Heart and Regenerative Medicine and.,Department of Basic Science Education, Virginia Tech Carilion School of Medicine and Research Institute, Roanoke, Virginia, USA.,Department of Biomedical Engineering and Mechanics.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| |
Collapse
|
48
|
Endothelial Ca 2+ Signaling and the Resistance to Anticancer Treatments: Partners in Crime. Int J Mol Sci 2018; 19:ijms19010217. [PMID: 29324706 PMCID: PMC5796166 DOI: 10.3390/ijms19010217] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 02/06/2023] Open
Abstract
Intracellular Ca2+ signaling drives angiogenesis and vasculogenesis by stimulating proliferation, migration, and tube formation in both vascular endothelial cells and endothelial colony forming cells (ECFCs), which represent the only endothelial precursor truly belonging to the endothelial phenotype. In addition, local Ca2+ signals at the endoplasmic reticulum (ER)-mitochondria interface regulate endothelial cell fate by stimulating survival or apoptosis depending on the extent of the mitochondrial Ca2+ increase. The present article aims at describing how remodeling of the endothelial Ca2+ toolkit contributes to establish intrinsic or acquired resistance to standard anti-cancer therapies. The endothelial Ca2+ toolkit undergoes a major alteration in tumor endothelial cells and tumor-associated ECFCs. These include changes in TRPV4 expression and increase in the expression of P2X7 receptors, Piezo2, Stim1, Orai1, TRPC1, TRPC5, Connexin 40 and dysregulation of the ER Ca2+ handling machinery. Additionally, remodeling of the endothelial Ca2+ toolkit could involve nicotinic acetylcholine receptors, gasotransmitters-gated channels, two-pore channels and Na⁺/H⁺ exchanger. Targeting the endothelial Ca2+ toolkit could represent an alternative adjuvant therapy to circumvent patients' resistance to current anti-cancer treatments.
Collapse
|
49
|
Han H, Du L, Cao Z, Zhang B, Zhou Q. Triptonide potently suppresses pancreatic cancer cell-mediated vasculogenic mimicry by inhibiting expression of VE-cadherin and chemokine ligand 2 genes. Eur J Pharmacol 2018; 818:593-603. [PMID: 29162433 DOI: 10.1016/j.ejphar.2017.11.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/10/2017] [Accepted: 11/13/2017] [Indexed: 02/06/2023]
Abstract
Various aggressive cancers, including pancreatic cancer, produce functional blood vessels by neovascularization. Tumor vasculogenic mimicry (VM) promotes cancer progression and is closely associated with the poor prognosis of the cancer patients. Therefore, tumor VM is a sensible target for novel anti-cancer drug discovery. However, there is a lack of effective anti-tumor VM drugs in the clinical setting. In this study, we aim to explore novel agents to effectually inhibit pancreatic cancer cell-mediated tumor VM for anti-cancer therapy. Pancreatic cancer cell lines Patu8988 and Panc1 were utilized as a model. A mouse model was used for in vitro capillary-like structure formation and in vivo Matrigel plug assays to evaluate the anti-tumor VM efficacy of a small molecule triptonide from traditional Chinese herbs. Various methods, including RT-PCR, immunohistochemical staining, and the luciferase gene transcription reporter system, were applied to study the mechanisms of triptonide-exerted anti-tumor VM. Triptonide effectively inhibited pancreatic cancer cell-formed capillary-like structures in vitro and blood vessels in vivo through suppressing pancreatic cancer cell migration, invasion, and VM via inhibiting expression of tumor VM master gene VE-cadherin and pro-migratory gene chemokine C-X-C motif ligand 2 (CXCL2), mainly via reduction of gene promoter activity. Triptonide potently suppresses pancreatic cancer cell-mediated VM by reducing tumor cell migration and invasion and inhibiting expression of VE-cadherin and CXCL2 genes. Our results provide a novel and potent anti-tumor VM drug candidate for further development of effective anti-pancreatic cancer therapy.
Collapse
Affiliation(s)
- Hongyan Han
- School of Biology and Basic Medical Sciences, Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China; 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Longsheng Du
- School of Biology and Basic Medical Sciences, Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China; 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Zhifei Cao
- School of Biology and Basic Medical Sciences, Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China; 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China.
| | - Bin Zhang
- School of Biology and Basic Medical Sciences, Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China; 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Quansheng Zhou
- School of Biology and Basic Medical Sciences, Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China; 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China.
| |
Collapse
|
50
|
Redundant angiogenic signaling and tumor drug resistance. Drug Resist Updat 2018; 36:47-76. [DOI: 10.1016/j.drup.2018.01.002] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/22/2017] [Accepted: 01/11/2018] [Indexed: 02/07/2023]
|