1
|
Malekan M, Haass NK, Rokni GR, Gholizadeh N, Ebrahimzadeh MA, Kazeminejad A. VEGF/VEGFR axis and its signaling in melanoma: Current knowledge toward therapeutic targeting agents and future perspectives. Life Sci 2024; 345:122563. [PMID: 38508233 DOI: 10.1016/j.lfs.2024.122563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/10/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024]
Abstract
Melanoma is responsible for most skin cancer-associated deaths globally. The progression of melanoma is influenced by a number of pathogenic processes. Understanding the VEGF/VEGFR axis, which includes VEGF-A, PlGF, VEGF-B, VEGF-C, and VEGF-D and their receptors, VEGFR-1, VEGFR-2, and VEGFR-3, is of great importance in melanoma due to its crucial role in angiogenesis. This axis generates multifactorial and complex cellular signaling, engaging the MAPK/ERK, PI3K/AKT, PKC, PLC-γ, and FAK signaling pathways. Melanoma cell growth and proliferation, migration and metastasis, survival, and acquired resistance to therapy are influenced by this axis. The VEGF/VEGFR axis was extensively examined for their potential as diagnostic/prognostic biomarkers in melanoma patients and results showed that VEGF overexpression can be associated with unfavorable prognosis, higher level of tumor invasion and poor response to therapy. MicroRNAs linking to the VEGF/VEGFR axis were identified and, in this review, divided into two categories according to their functions, some of them promote melanoma angiogenesis (promotive group) and some restrict melanoma angiogenesis (protective group). In addition, the approach of treating melanoma by targeting the VEGF/VEGFR axis has garnered significant interest among researchers. These agents can be divided into two main groups: anti-VEGF and VEGFR inhibitors. These therapeutic options may be a prominent step along with the modern targeting and immune therapies for better coverage of pathological processes leading to melanoma progression and therapy resistance.
Collapse
Affiliation(s)
- Mohammad Malekan
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | | | - Ghasem Rahmatpour Rokni
- Department of Dermatology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nasim Gholizadeh
- Department of Dermatology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Ali Ebrahimzadeh
- Pharmaceutical Sciences Research Center, School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Armaghan Kazeminejad
- Department of Dermatology, Antimicrobial Resistance Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences,Sari, Iran
| |
Collapse
|
2
|
García-Hevia L, Soltani R, González J, Chaloin O, Ménard-Moyon C, Bianco A, L. Fanarraga M. Carbon nanotubes targeted to the tumor microenvironment inhibit metastasis in a preclinical model of melanoma. Bioact Mater 2024; 34:237-247. [PMID: 38223536 PMCID: PMC10787223 DOI: 10.1016/j.bioactmat.2023.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/16/2024] Open
Abstract
Despite notable progress in cancer therapy, metastatic diseases continue to be the primary cause of cancer-related mortality. Multi-walled carbon nanotubes (MWCNTs) can enter tissues and cells and interfere with the dynamics of the cytoskeletal nanofilaments biomimetically. This endows them with intrinsic anti-tumoral effects comparable to those of microtubule-binding chemotherapies such as Taxol®. In this study, our focus was on exploring the potential of oxidized MWCNTs in selectively targeting the vascular endothelial growth factor receptor (VEGFR). Our objective was to evaluate their effectiveness in inhibiting metastatic growth by inducing anti-proliferative, anti-migratory, and cytotoxic effects on both cancer and tumor microenvironment cells. Our findings demonstrated a significant reduction of over 80 % in malignant melanoma lung metastases and a substantial enhancement in overall animal welfare following intravenous administration of the targeted biodegradable MWCNTs. Furthermore, the combination of these nanomaterials with the conventional chemotherapy agent Taxol® yielded a remarkable 90 % increase in the antimetastatic effect. These results highlight the promising potential of this combined therapeutic approach against metastatic disease and are of paramount importance as metastasis is responsible for nearly 60,000 deaths each year.
Collapse
Affiliation(s)
- Lorena García-Hevia
- The Nanomedicine Group, Universidad de Cantabria-IDIVAL, Avda Herrera Oria s/n, 39011, Santander, Spain
| | - Rym Soltani
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000, Strasbourg, France
| | - Jesús González
- The Nanomedicine Group, Universidad de Cantabria-IDIVAL, Avda Herrera Oria s/n, 39011, Santander, Spain
| | - Olivier Chaloin
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000, Strasbourg, France
| | - Cécilia Ménard-Moyon
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000, Strasbourg, France
| | - Alberto Bianco
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000, Strasbourg, France
| | - Mónica L. Fanarraga
- The Nanomedicine Group, Universidad de Cantabria-IDIVAL, Avda Herrera Oria s/n, 39011, Santander, Spain
| |
Collapse
|
3
|
Hsu MJ, Chen HK, Lien JC, Huang YH, Huang SW. Suppressing VEGF-A/VEGFR-2 Signaling Contributes to the Anti-Angiogenic Effects of PPE8, a Novel Naphthoquinone-Based Compound. Cells 2022; 11:cells11132114. [PMID: 35805198 PMCID: PMC9266117 DOI: 10.3390/cells11132114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/30/2022] [Accepted: 07/02/2022] [Indexed: 02/05/2023] Open
Abstract
Natural naphthoquinones and their derivatives exhibit a broad spectrum of pharmacological activities and have thus attracted much attention in modern drug discovery. However, it remains unclear whether naphthoquinones are potential drug candidates for anti-angiogenic agents. The aim of this study was to evaluate the anti-angiogenic properties of a novel naphthoquinone derivative, PPE8, and explore its underlying mechanisms. Determined by various assays including BrdU, migration, invasion, and tube formation analyses, PPE8 treatment resulted in the reduction of VEGF-A-induced proliferation, migration, and invasion, as well as tube formation in human umbilical vein endothelial cells (HUVECs). We also used an aorta ring sprouting assay, Matrigel plug assay, and immunoblotting analysis to examine PPE8’s ex vivo and in vivo anti-angiogenic activities and its actions on VEGF-A signaling. It has been revealed that PPE8 inhibited VEGF-A-induced micro vessel sprouting and was capable of suppressing angiogenesis in in vivo models. In addition, PPE8 inhibited VEGF receptor (VEGFR)-2, Src, FAK, ERK1/2, or AKT phosphorylation in HUVECs exposed to VEGF-A, and it also showed significant decline in xenograft tumor growth in vivo. Taken together, these observations indicated that PPE8 may target VEGF-A–VEGFR-2 signaling to reduce angiogenesis. It also supports the role of PPE8 as a potential drug candidate for the development of therapeutic agents in the treatment of angiogenesis-related diseases including cancer.
Collapse
Affiliation(s)
- Ming-Jen Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Han-Kun Chen
- Department of General Surgery, Chi Mei Medical Center, Tainan 71067, Taiwan;
| | - Jin-Cherng Lien
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan;
- Department of Medical Research, Hospital of China Medical University, Taichung 40402, Taiwan
| | - Yu-Han Huang
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Shiu-Wen Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Medical Research, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Research Center of Thoracic Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Correspondence: ; Tel.: +886-2-27361661 (ext. 3198)
| |
Collapse
|
4
|
Hsu MJ, Chen HK, Chen CY, Lien JC, Gao JY, Huang YH, Hsu JBK, Lee GA, Huang SW. Anti-Angiogenetic and Anti-Lymphangiogenic Effects of a Novel 2-Aminobenzimidazole Derivative, MFB. Front Oncol 2022; 12:862326. [PMID: 35795066 PMCID: PMC9251317 DOI: 10.3389/fonc.2022.862326] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/24/2022] [Indexed: 12/02/2022] Open
Abstract
Background and Purpose Benzimidazoles have attracted much attention over the last few decades due to their broad-spectrum pharmacological properties. Increasing evidence is showing the potential use of benzimidazoles as anti-angiogenic agents, although the mechanisms that impact angiogenesis remain to be fully defined. In this study, we aim to investigate the anti-angiogenic mechanisms of MFB, a novel 2-aminobenzimidazole derivative, to develop a novel angiogenesis inhibitor. Experimental Approach MTT, BrdU, migration and invasion assays, and immunoblotting were employed to examine MFB’s effects on vascular endothelial growth factor (VEGF)-induced endothelial cell proliferation, migration, invasion, as well as signaling molecules activation. The anti-angiogenic effects of MFB were analyzed by tube formation, aorta ring sprouting, and matrigel plug assays. We also used a mouse model of lung metastasis to determine the MFB’s anti-metastatic effects. Key Results MFB suppressed cell proliferation, migration, invasion, and endothelial tube formation of VEGF-A-stimulated human umbilical vascular endothelial cells (HUVECs) or VEGF-C-stimulated lymphatic endothelial cells (LECs). MFB suppressed VEGF-A and VEGF-C signaling in HUVECs or LECs. In addition, MFB reduced VEGF-A- or tumor cells-induced neovascularization in vivo. MFB also diminished B16F10 melanoma lung metastasis. The molecular docking results further showed that MFB may bind to VEGFR-2 rather than VEGF-A with high affinity. Conclusions and Implications These observations indicated that MFB may target VEGF/VEGFR signaling to suppress angiogenesis and lymphangiogenesis. It also supports the role of MFB as a potential lead in developing novel agents for the treatment of angiogenesis- or lymphangiogenesis-associated diseases and cancer.
Collapse
Affiliation(s)
- Ming-Jen Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Han-Kun Chen
- Department of General Surgery, Chi Mei Medical Center, Tainan, Taiwan
| | - Cheng-Yu Chen
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Radiology, National Defense Medical Center, Taipei, Taiwan
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jin-Cherng Lien
- School of Pharmacy, China Medical University, Taichung, Taiwan
- Department of Medical Research, Hospital of China Medical University, Taichung, Taiwan
| | - Jing-Yan Gao
- School of Pharmacy, China Medical University, Taichung, Taiwan
- Department of Medical Research, Hospital of China Medical University, Taichung, Taiwan
| | - Yu-Han Huang
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA, United States
| | - Justin Bo-Kai Hsu
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Medical Research; Research Center of Thoracic Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Gilbert Aaron Lee
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Medical Research; Research Center of Thoracic Medicine, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shiu-Wen Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Medical Research; Research Center of Thoracic Medicine, Taipei Medical University Hospital, Taipei, Taiwan
- Research Center of Thoracic Medicine, Taipei Medical University Hospital, Taipei, Taiwan
- *Correspondence: Shiu-Wen Huang,
| |
Collapse
|
5
|
Abstract
Melanoma is a relentless type of skin cancer which involves myriad signaling pathways which regulate many cellular processes. This makes melanoma difficult to treat, especially when identified late. At present, therapeutics include chemotherapy, surgical resection, biochemotherapy, immunotherapy, photodynamic and targeted approaches. These interventions are usually administered as either a single-drug or in combination, based on tumor location, stage, and patients' overall health condition. However, treatment efficacy generally decreases as patients develop treatment resistance. Genetic profiling of melanocytes and the discovery of novel molecular factors involved in the pathogenesis of melanoma have helped to identify new therapeutic targets. In this literature review, we examine several newly approved therapies, and briefly describe several therapies being assessed for melanoma. The goal is to provide a comprehensive overview of recent developments and to consider future directions in the field of melanoma.
Collapse
Affiliation(s)
- Pavan Kumar Dhanyamraju
- Department of Pediatrics and Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Pavan Kumar Dhanyamraju, Department of Pediatrics and Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA17033, USA. Tel: +1-6096474712, E-mail:
| | - Trupti N. Patel
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Vellore, Tamil Nadu 632014, India
| |
Collapse
|
6
|
Tie-2, G-CSF, and Leptin as Promising Diagnostic Biomarkers for Endometrial Cancer: A Pilot Study. J Clin Med 2021; 10:jcm10040765. [PMID: 33671851 PMCID: PMC7918088 DOI: 10.3390/jcm10040765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/28/2021] [Accepted: 02/03/2021] [Indexed: 12/23/2022] Open
Abstract
Preoperative determination of the extent of endometrial cancer (EC) would avoid the complications associated with radical surgery. Screening of patients' plasma biomarkers might enable a more precise diagnosis of EC and a tailored treatment approach. This prospective case-control monocentric pilot study included 76 postmenopausal women (38 endometrioid EC patients and 38 control patients with benign gynecological conditions), and 37 angiogenic factors (AFs) were investigated as potential biomarkers for EC. AF concentrations in preoperative plasma samples were measured using Luminex xMAP™ multiplexing technology. The plasma levels of sTie-2 and G-CSF were significantly lower in EC compared to control patients, whereas the plasma levels of leptin were significantly higher in EC patients. Neuropilin-1 plasma levels were significantly higher in patients with type 2 EC (grade 3) compared to patients with lower grade cancer or controls. Follistatin levels were significantly higher in patients with lymphovascular invasion, and IL-8 plasma levels were significantly higher in patients with metastases. If validated, the plasma concentrations of the indicated AFs could represent an important additional diagnostic tool for the early detection and characterization of EC. This could guide the decision-making on the extent of surgery. Further studies with larger patient numbers are currently ongoing.
Collapse
|
7
|
Canè S, Van Snick J, Uyttenhove C, Pilotte L, Van den Eynde BJ. TGFβ1 neutralization displays therapeutic efficacy through both an immunomodulatory and a non-immune tumor-intrinsic mechanism. J Immunother Cancer 2021; 9:e001798. [PMID: 33637600 PMCID: PMC7919595 DOI: 10.1136/jitc-2020-001798] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Transforming growth factor-β (TGFβ) is emerging as a promising target for cancer therapy, given its ability to promote progression of advanced tumors and to suppress anti-tumor immune responses. However, TGFβ also plays multiple roles in normal tissues, particularly during organogenesis, raising toxicity concerns about TGFβ blockade. Dose-limiting cardiovascular toxicity was observed, possibly due to the blockade of all three TGFβ isoforms. The dominant isoform in tumors is TGFβ1, while TGFβ2 and TGFβ3 seem to be more involved in cardiovascular development. Recent data indicated that selective targeting of TGFβ1 promoted the efficacy of checkpoint inhibitor anti-PD1 in transplanted preclinical tumor models, without cardiovascular toxicity. METHODS To further explore the therapeutic potential of isoform-specific TGFβ blockade, we developed neutralizing mAbs targeting mature TGFβ1 or TGFβ3, and tested them, in parallel with anti-panTGFβ mAb 1D11, in two preclinical models: the transplanted colon cancer model CT26, and the autochthonous melanoma model TiRP. RESULTS We observed that the blockade of TGFβ1, but not that of TGFβ3, increased the efficacy of a prophylactic cellular vaccine against colon cancer CT26. This effect was similar to pan-TGFβ blockade, and was associated with increased infiltration of activated CD8 T cells in the tumor, and reduced levels of regulatory T cells and myeloid-derived suppressor cells. In contrast, in the autochthonous TiRP melanoma model, we observed therapeutic efficacy of the TGFβ1-specific mAb as a single agent, while the TGFβ3 mAb was inactive. In this model, the anti-tumor effect of TGFβ1 blockade was tumor intrinsic rather than immune mediated, as it was also observed in T-cell depleted mice. Mechanistically, TGFβ1 blockade increased mouse survival by delaying the phenotype switch, akin to epithelial-to-mesenchymal transition (EMT), which transforms initially pigmented tumors into highly aggressive unpigmented tumors. CONCLUSIONS Our results confirm TGFβ1 as the relevant isoform to target for cancer therapy, not only in combination with checkpoint inhibitors, but also with other immunotherapies such as cancer vaccines. Moreover, TGFβ1 blockade can also act as a monotherapy, through a tumor-intrinsic effect blocking the EMT-like transition. Because human melanomas that resist therapy often express a gene signature that links TGFβ1 with EMT-related genes, these results support the clinical development of TGFβ1-specific mAbs in melanoma.
Collapse
Affiliation(s)
- Stefania Canè
- Ludwig Institute for Cancer Research, De Duve Institute, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
- Department of Medicine, Immunology Section, University of Verona, Verona, Italy
| | - Jacques Van Snick
- Ludwig Institute for Cancer Research, De Duve Institute, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
| | - Catherine Uyttenhove
- Ludwig Institute for Cancer Research, De Duve Institute, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
| | - Luc Pilotte
- Ludwig Institute for Cancer Research, De Duve Institute, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
- WELBIO, UCLouvain, Brussels, Belgium
| | - Benoit J Van den Eynde
- Ludwig Institute for Cancer Research, De Duve Institute, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
- WELBIO, UCLouvain, Brussels, Belgium
| |
Collapse
|
8
|
Yazdani M, Jaafari MR, Verdi J, Alani B, Noureddini M, Badiee A. Ex vivo-generated dendritic cell-based vaccines in melanoma: the role of nanoparticulate delivery systems. Immunotherapy 2020; 12:333-349. [DOI: 10.2217/imt-2019-0173] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Melanoma is a poor immunogenic cancer and many treatment strategies have been used to enhance specific or nonspecific immunity against it. Dendritic cell (DC)-based cancer vaccine is the most effective therapies that have been used so far. Meanwhile, the efficacy of DC-based immunotherapy relies on critical factors relating to DCs such as the state of maturation and proper delivery of antigens. In this regard, the use of nanoparticulate delivery systems for effective delivery of antigen to ex vivo-generated DC-based vaccines that also poses adjuvanticity would be an ideal approach. In this review article, we attempt to summarize the role of different types of nanoparticulate antigen delivery systems used in the development of ex vivo-generated DC-based vaccines against melanoma and describe their adjuvanticity in mediation of DC maturation, cytoplasmic presentation of antigens to MHC class I molecules, which led to potent antigen-specific immune responses. As were represented, cationic liposomes were the most used approach, which suggest its potential applicability as delivery systems for further experiments in combination with either adjuvants or monoclonal antibodies.
Collapse
Affiliation(s)
- Mona Yazdani
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan 91778-99191, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
| | - Mahmoud Reza Jaafari
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
| | - Javad Verdi
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan 91778-99191, Iran
| | - Behrang Alani
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan 91778-99191, Iran
| | - Mahdi Noureddini
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan 91778-99191, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
| |
Collapse
|
9
|
Jackett LA, Scolyer RA. A Review of Key Biological and Molecular Events Underpinning Transformation of Melanocytes to Primary and Metastatic Melanoma. Cancers (Basel) 2019; 11:cancers11122041. [PMID: 31861163 PMCID: PMC6966527 DOI: 10.3390/cancers11122041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/02/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023] Open
Abstract
Melanoma is a major public health concern that is responsible for significant morbidity and mortality, particularly in countries such as New Zealand and Australia where it is the commonest cause of cancer death in young adults. Until recently, there were no effective drug therapies for patients with advanced melanoma however significant advances in our understanding of the biological and molecular basis of melanoma in recent decades have led to the development of revolutionary treatments, including targeted molecular therapy and immunotherapy. This review summarizes our current understanding of the key events in the pathway of melanomagenesis and discusses the role of genomic analysis as a potential tool for improved diagnostic evaluation, prognostication and treatment strategies. Ultimately, it is hoped that a continued deeper understanding of the mechanisms of melanomagenesis will lead to the development of even more effective treatments that continue to provide better outcomes for patients with melanoma.
Collapse
Affiliation(s)
- Louise A. Jackett
- Melanoma Institute Australia, 2065 Sydney, Australia;
- Sydney Medical School, The University of Sydney, 2050 Sydney, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, 2050 Sydney, Australia
- Department of Anatomical Pathology, Austin Hospital, 3084 Melbourne, Australia
| | - Richard A. Scolyer
- Melanoma Institute Australia, 2065 Sydney, Australia;
- Sydney Medical School, The University of Sydney, 2050 Sydney, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, 2050 Sydney, Australia
- Correspondence: ; Tel.: +61-299117200; Fax: +61-299549290
| |
Collapse
|
10
|
Lien J, Chung C, Huang T, Chang T, Chen K, Gao G, Hsu M, Huang S. A novel 2-aminobenzimidazole-based compound Jzu 17 exhibits anti-angiogenesis effects by targeting VEGFR-2 signalling. Br J Pharmacol 2019; 176:4034-4049. [PMID: 31368127 PMCID: PMC6811776 DOI: 10.1111/bph.14813] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Recent development in drug discovery have shown benzimidazole to be an important pharmacophore,. Benzimidazole derivatives exhibit broad-spectrum pharmacological properties including anti-microbial, anti-diabetic and anti-tumour activity. However, whether benzimidazole derivatives are effective in suppressing angiogenesis and its underlying mechanisms remain incompletely understood. In this study, we aim to characterize the anti-angiogenic mechanisms of a novel 2-aminobenzimidazole-based compound, Jzu 17, in an effort to develop novel angiogenesis inhibitor. EXPERIMENTAL APPROACH Effects of Jzu 17 on endothelial cell proliferation, migration, invasion, and activation of signalling molecules induced by VEGF-A, were analysed by immunoblotting, MTT, BrdU, migration, and invasion assays. We performed tube formation assay, aorta ring sprouting assay, matrigel plug assay, and a mouse model of metastasis to evaluate ex vivo and in vivo anti-angiogenic effects of Jzu 17. KEY RESULTS Jzu 17 inhibited VEGF-A-induced cell proliferation, migration, invasion, and endothelial tube formation of HUVECs. Jzu 17 suppressed VEGF-A-induced microvessel sprouting ex vivo and attenuated VEGF-A- or tumour cell-induced neovascularization in vivo. Jzu 17 also reduced B16F10 melanoma lung metastasis. In addition, Jzu 17 inhibited the phosphorylation of VEGFR-2 and its downstream signalling molecules in VEGF-A-stimulated HUVECs. Results from computer modelling further showed that Jzu 17 binds to VEGFR-2 with high affinity. CONCLUSIONS AND IMPLICATIONS Jzu 17 may inhibit endothelial remodelling and suppress angiogenesis through targeting VEGF-A-VEGFR-2 signalling. These results also suggest Jzu 17 as a potential lead compound and warrant the clinical development of similar agents in the treatment of cancer and angiogenesis-related diseases.
Collapse
Affiliation(s)
- Jin‐Cherng Lien
- School of PharmacyChina Medical UniversityTaichungTaiwan
- Department of Medical ResearchHospital of China Medical UniversityTaichungTaiwan
| | - Chi‐Li Chung
- Division of Pulmonary Medicine, Department of Internal MedicineTaipei Medical University HospitalTaipeiTaiwan
- Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine and School of Respiratory Therapy, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Tur‐Fu Huang
- Graduate Institute of Pharmacology, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| | | | | | - Ging‐Yan Gao
- School of PharmacyChina Medical UniversityTaichungTaiwan
| | - Ming‐Jen Hsu
- Department of Pharmacology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Shiu‐Wen Huang
- Department of Pharmacology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Department of Medical ResearchTaipei Medical University HospitalTaipeiTaiwan
| |
Collapse
|
11
|
Monteiro AC, Muenzner JK, Andrade F, Rius FE, Ostalecki C, Geppert CI, Agaimy A, Hartmann A, Fujita A, Schneider-Stock R, Jasiulionis MG. Gene expression and promoter methylation of angiogenic and lymphangiogenic factors as prognostic markers in melanoma. Mol Oncol 2019; 13:1433-1449. [PMID: 31069961 PMCID: PMC6547615 DOI: 10.1002/1878-0261.12501] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/18/2019] [Accepted: 05/08/2019] [Indexed: 01/06/2023] Open
Abstract
The high mortality rate of melanoma is broadly associated with its metastatic potential. Tumor cell dissemination is strictly dependent on vascularization; therefore, angiogenesis and lymphangiogenesis play an essential role in metastasis. Hence, a better understanding of the players of tumor vascularization and establishing them as new molecular biomarkers might help to overcome the poor prognosis of melanoma patients. Here, we further characterized a linear murine model of melanoma progression and showed that the aggressiveness of melanoma cells is closely associated with high expression of angiogenic factors, such as Vegfc, Angpt2, and Six1, and that blockade of the vascular endothelial growth factor pathway by the inhibitor axitinib abrogates their tumorigenic potential in vitro and in the in vivo chicken chorioallantoic membrane assay. Furthermore, analysis of The Cancer Genome Atlas data revealed that the expression of the angiogenic factor ANGPT2 (P‐value = 0.044) and the lymphangiogenic receptor VEGFR‐3 (P‐value = 0.002) were independent prognostic factors of overall survival in melanoma patients. Enhanced reduced representation bisulfite sequencing‐based methylome profiling revealed for the first time a link between abnormal VEGFC, ANGPT2, and SIX1 gene expression and promoter hypomethylation in melanoma cells. In patients, VEGFC (P‐value = 0.031), ANGPT2 (P‐value < 0.001), and SIX1 (P‐value = 0.009) promoter hypomethylation were independent prognostic factors of shorter overall survival. Hence, our data suggest that these angio‐ and lymphangiogenesis factors are potential biomarkers of melanoma prognosis. Moreover, these findings strongly support the applicability of our melanoma progression model to unravel new biomarkers for this aggressive human disease.
Collapse
Affiliation(s)
- Ana Carolina Monteiro
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Brazil.,Department of Experimental Tumor Pathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Julienne K Muenzner
- Department of Experimental Tumor Pathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Fernando Andrade
- Department of Computer Science, Institute of Mathematics and Statistics, Universidade de São Paulo, Brazil
| | - Flávia Eichemberger Rius
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Brazil
| | - Christian Ostalecki
- Department of Dermatology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Germany
| | - Carol I Geppert
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Abbas Agaimy
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Arndt Hartmann
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - André Fujita
- Department of Computer Science, Institute of Mathematics and Statistics, Universidade de São Paulo, Brazil
| | - Regine Schneider-Stock
- Department of Experimental Tumor Pathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | | |
Collapse
|
12
|
Abstract
The Hippo signaling pathway is involved in tissue size regulation and tumorigenesis. Genetic deletion or aberrant expression of some Hippo pathway genes lead to enhanced cell proliferation, tumorigenesis, and cancer metastasis. Recently, multiple studies have identified a wide range of upstream regulators of the Hippo pathway, including mechanical cues and ligands of G protein-coupled receptors (GPCRs). Through the activation related G proteins and possibly rearrangements of actin cytoskeleton, GPCR signaling can potently modulate the phosphorylation states and activity of YAP and TAZ, two homologous oncogenic transcriptional co-activators, and major effectors of the Hippo pathway. Herein, we summarize the network, regulation, and functions of GPCR-Hippo signaling, and we will also discuss potential anti-cancer therapies targeting GPCR-YAP signaling.
Collapse
|
13
|
Tarhini AA, Frankel P, Ruel C, Ernstoff MS, Kuzel TM, Logan TF, Khushalani NI, Tawbi HA, Margolin KA, Awasthi S, Butterfield LH, McDermott D, Chen A, Lara PN, Kirkwood JM. NCI 8628: A randomized phase 2 study of ziv-aflibercept and high-dose interleukin 2 or high-dose interleukin 2 alone for inoperable stage III or IV melanoma. Cancer 2018; 124:4332-4341. [PMID: 30303516 PMCID: PMC6504933 DOI: 10.1002/cncr.31734] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 08/22/2016] [Accepted: 10/31/2016] [Indexed: 11/09/2022]
Abstract
BACKGROUND Interleukin 2 (IL-2) is a growth factor for T and natural killer cells, promotes proinflammatory cytokines, and can lead to durable responses in patients with melanoma. Vascular endothelial growth factor (VEGF) promotes angiogenesis and modulates host innate and adaptive immunity. High VEGF levels were found to be associated with nonresponse to IL-2. Ziv-aflibercept may deplete VEGF and thereby enhance antitumor T-cell responses, thus supporting a combination immunotherapeutic strategy with IL-2. METHODS NCI 8628 was a phase 2 trial of ziv-aflibercept and IL-2 (arm A) versus IL-2 alone (arm B) randomized at 2:1, respectively. Eligible patients had inoperable American Joint Committee on Cancer stage III or stage IV melanoma. The primary endpoint was progression-free survival (PFS). RESULTS A total of 89 patients were enrolled and 84 patients were treated. The median follow-up was 41.4 months. Among treated patients (55 patients in arm A and 29 patients in arm B), PFS was significantly improved in favor of arm A, with a median of 6.9 months (95% confidence interval [95% CI], 4.1-8.7 months) versus 2.3 months (95% CI, 1.6-3.5 months) (P<.001). No significant difference was noted with regard to overall survival, with a median of 26.9 months (95% CI, 14.4-63.6 months) for arm A and 24.2 months (95% CI, 11.3-36.4 months) for arm B. The response rate (according to Response Evaluation Criteria In Solid Tumors [RECIST]) was 22% in arm A (4 complete responses [CRs] and 8 partial responses [PRs]) and 17% in arm B (1 CR and 4 PRs). Stable disease or PR or CR was noted in 65% of patients in arm A and 48% of patients in arm B. The combination was found to be superior to monotherapy in patients with high and low levels of serum VEGF and VEGF receptor 2. Adverse events were consistent with the expected profiles of monotherapy with IL-2 and ziv-aflibercept. CONCLUSIONS Ziv-aflibercept and IL-2 were found to significantly improve PFS compared with IL-2 alone, thereby meeting the primary endpoint of the current study. These findings support further study of immunotherapeutic combination strategies involving VEGF inhibitors.
Collapse
Affiliation(s)
- Ahmad A. Tarhini
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Cleveland Clinic and Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Paul Frankel
- City of Hope National Medical Center, Duarte, CA, USA
| | | | | | | | | | | | - Hussein A. Tawbi
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Lisa H. Butterfield
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Departments of Medicine, Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Alice Chen
- Cancer Therapy Evaluation Program (CTEP), National Cancer Institute, Bethesda, MD, USA
| | - Primo N. Lara
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | | |
Collapse
|
14
|
Abstract
Melanoma represents the most aggressive and the deadliest form of skin cancer. Current therapeutic approaches include surgical resection, chemotherapy, photodynamic therapy, immunotherapy, biochemotherapy, and targeted therapy. The therapeutic strategy can include single agents or combined therapies, depending on the patient’s health, stage, and location of the tumor. The efficiency of these treatments can be decreased due to the development of diverse resistance mechanisms. New therapeutic targets have emerged from studies of the genetic profile of melanocytes and from the identification of molecular factors involved in the pathogenesis of the malignant transformation. In this review, we aim to survey therapies approved and under evaluation for melanoma treatment and relevant research on the molecular mechanisms underlying melanomagenesis.
Collapse
Affiliation(s)
- Beatriz Domingues
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Faculty of Sciences, University of Porto, Porto, Portugal
| | - José Manuel Lopes
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Department of Pathology, Hospital S João, Porto, Portugal.,Department of Pathology, Medical Faculty, University of Porto, Porto, Portugal
| | - Paula Soares
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Department of Pathology, Medical Faculty, University of Porto, Porto, Portugal
| | - Helena Pópulo
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
15
|
The Potential of Angiogenin as a Serum Biomarker for Diseases: Systematic Review and Meta-Analysis. DISEASE MARKERS 2018; 2018:1984718. [PMID: 29736193 PMCID: PMC5875026 DOI: 10.1155/2018/1984718] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/02/2018] [Accepted: 01/11/2018] [Indexed: 12/19/2022]
Abstract
Background Angiogenin (ANG) is a multifunctional angiogenic protein that participates in both normal development and diseases. Abnormal serum ANG levels are commonly reported in various diseases. However, whether ANG can serve as a diagnostic or prognostic marker for different diseases remains a matter of debate. Methods Here, we performed a systematic review and meta-analysis of the literature utilizing PubMed, Web of Science, and Scopus search engines to identify all publications comparing plasma or serum ANG levels between patients with different diseases and healthy controls, as were studies evaluating circulating ANG levels in healthy populations, pregnant women, or other demographic populations. Results This study demonstrated that the serum ANG concentration in healthy populations was 336.14 ± 142.83 ng/ml and remained relatively stable in different populations and regions. We noted no significant differences in serum ANG levels between patients and healthy controls, except in cases in which patients suffered from cancer or cardiovascular diseases. The serum ANG concentrations were significantly higher in patients who developed colorectal cancer, acute myeloid leukemia, multiple myeloma, myelodysplastic syndromes, and heart failure than those in healthy controls. Conclusion ANG has the potential of being a serum biomarker for cancers and cardiovascular diseases.
Collapse
|
16
|
Daud A, Kluger HM, Kurzrock R, Schimmoller F, Weitzman AL, Samuel TA, Moussa AH, Gordon MS, Shapiro GI. Phase II randomised discontinuation trial of the MET/VEGF receptor inhibitor cabozantinib in metastatic melanoma. Br J Cancer 2017; 116:432-440. [PMID: 28103611 PMCID: PMC5318966 DOI: 10.1038/bjc.2016.419] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 10/20/2016] [Accepted: 11/14/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND A phase II randomised discontinuation trial assessed cabozantinib (XL184), an orally bioavailable inhibitor of tyrosine kinases including VEGF receptors, MET, and AXL, in a cohort of patients with metastatic melanoma. METHODS Patients received cabozantinib 100 mg daily during a 12-week lead-in. Patients with stable disease (SD) per Response Evaluation Criteria in Solid Tumours (RECIST) at week 12 were randomised to cabozantinib or placebo. Primary endpoints were objective response rate (ORR) at week 12 and postrandomisation progression-free survival (PFS). RESULTS Seventy-seven patients were enroled (62% cutaneous, 30% uveal, and 8% mucosal). At week 12, the ORR was 5%; 39% of patients had SD. During the lead-in phase, reduction in target lesions from baseline was seen in 55% of evaluable patients overall and in 59% of evaluable patients with uveal melanoma. Median PFS after randomisation was 4.1 months with cabozantinib and 2.8 months with placebo (hazard ratio of 0.59; P=0.284). Median PFS from study day 1 was 3.8 months, 6-month PFS was 33%, and median overall survival was 9.4 months. The most common grade 3/4 adverse events were fatigue (14%), hypertension (10%), and abdominal pain (8%). One treatment-related death was reported from peritonitis due to diverticular perforation. CONCLUSIONS Cabozantinib has clinical activity in patients with metastatic melanoma, including uveal melanoma. Further clinical investigation is warranted.
Collapse
Affiliation(s)
- Adil Daud
- University of California, San Francisco Medical Center at Parnassus, 1600 Divisadero Street, MZ Bldg A, San Francisco, CA 94115, USA
| | - Harriet M Kluger
- Yale Cancer Center, Yale University School of Medicine, 333 Cedar Street, PO Box 208028, New Haven, CT 06520-8028, USA
| | - Razelle Kurzrock
- University of California, San Diego Moores Cancer Center, 3855 Health Sciences Drive, MC #0658, La Jolla, CA 92093-0658, USA
| | | | - Aaron L Weitzman
- Exelixis Inc., 210 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Thomas A Samuel
- Department of Hematology/Oncology, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd, Weston, FL 33331, USA
| | - Ali H Moussa
- Cancer Care Associates, 1810 E 15th Street, Tulsa, OK 74104, USA
| | - Michael S Gordon
- Pinnacle Oncology Hematology, 9055 E. Del Camino, Suite 100, Scottsdale, AZ 85258, USA
| | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Early Drug Development Center, 450 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
17
|
Das AM, Pescatori M, Vermeulen CE, Rens JAP, Seynhaeve ALB, Koning GA, Eggermont AMM, Ten Hagen TLM. Melanomas prevent endothelial cell death under restrictive culture conditions by signaling through AKT and p38 MAPK/ ERK-1/2 cascades. Oncoimmunology 2016; 5:e1219826. [PMID: 27853641 PMCID: PMC5087299 DOI: 10.1080/2162402x.2016.1219826] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 12/18/2022] Open
Abstract
Although melanoma progression and staging is clinically well characterized, a large variation is observed in pathogenesis, progression, and therapeutic responses. Clearly, intrinsic characteristics of melanoma cells contribute to this variety. An important factor, in both progression of the disease and response to therapy, is the tumor-associated vasculature. We postulate that melanoma cells communicate with endothelial cells (ECs) in order to establish a functional and supportive blood supply. We investigated the angiogenic potential of human melanoma cell lines by monitoring the survival of ECs upon exposure to melanoma conditioned medium (CM), under restrictive conditions. We observed long-term (up to 72 h) EC survival under hypoxic conditions upon treatment with all melanoma CMs. No such survival effect was observed with the CM of melanocytes. The CM of pancreatic and breast tumor cell lines did not show a long-term survival effect, suggesting that the survival factor is specific to melanoma cells. Furthermore, all size fractions (up to < 1 kDa) of the melanoma CM induced long-term survival of ECs. The survival effect observed by the < 1 kDa fraction excludes known pro-angiogenic factors. Heat inactivation and enzymatic digestion of the CM did not inactivate the survival factor. Global gene expression and pathway analysis suggest that this effect is mediated in part via the AKT and p38 MAPK/ ERK-1/2 signaling axis. Taken together, these data indicate the production of (a) survival factor/s (< 1 kDa) by melanoma cell lines, which enables long-term survival of ECs and promotes melanoma-induced angiogenesis.
Collapse
Affiliation(s)
- Asha M Das
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center , Rotterdam, the Netherlands
| | - Mario Pescatori
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center , Rotterdam, the Netherlands
| | - Cindy E Vermeulen
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center , Rotterdam, the Netherlands
| | - Joost A P Rens
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center , Rotterdam, the Netherlands
| | - Ann L B Seynhaeve
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center , Rotterdam, the Netherlands
| | - Gerben A Koning
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center , Rotterdam, the Netherlands
| | - Alexander M M Eggermont
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center, Rotterdam, the Netherlands; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - Timo L M Ten Hagen
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus Medical Center , Rotterdam, the Netherlands
| |
Collapse
|
18
|
Shi J, Lu Y, Wei P. Xiaotan Sanjie decoction inhibits angiogenesis in gastric cancer through Interleukin-8-linked regulation of the vascular endothelial growth factor pathway. JOURNAL OF ETHNOPHARMACOLOGY 2016; 189:230-237. [PMID: 27224240 DOI: 10.1016/j.jep.2016.05.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 04/30/2016] [Accepted: 05/16/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Interleukin-8 (IL-8) as a pro-angiogenic factor is strongly associated with gastric cancer metastasis. Xiaotan Sanjie (XTSJ) decoction is an empirical compound prescription based on the phlegm theory of traditional Chinese medicine. Previous studies have shown that XTSJ decoction decreases IL-8 level and formation of vasculogenic mimicry of gastric cancer. AIM OF THE STUDY To investigate the link between Xiaotan Sanjie (XTSJ) decoction and IL-8 regulation in the angiogenesis of gastric cancer. MATERIALS AND METHODS Human umbilical vein endothelial cells (HUVECs) were co-cultured with SGC-7901 human gastric cancer cells and exposed to serum samples containing XTSJ decoction and/or IL-8 (1.0ng/mL). The canalization and migration capacities were evaluated by tube formation and transwell migration assay. Protein (immunofluorescence and Western blot) and mRNA (qPCR) expressions were measured in 24-h-cultured HUVECs for vascular endothelial growth factor-A (VEGF-A), vascular endothelial growth factor receptor (VEGFR)-1, and VEGFR-2. RESULTS IL-8 significantly promoted and XTSJ decoction inhibited HUVEC tube formation and migration. Links between IL-8 regulation and XTSJ decoction were found in tube formation and migration assays. IL-8 upregulated and XTSJ decoction downregulated VEGF-A, VEGFR-1, and VEGFR-2 protein levels. XTSJ decoction inhibited IL-8-induced VEGF-A and VEGFR-1 protein expressions. Similarly, IL-8 promoted VEGF-A, VEGFR-1, and VEGFR-2 mRNA levels; however, XTSJ decoction inhibited only VEGF-A mRNA. Interestingly, XTSJ decoction inhibited IL-8-induced VEGFR-1 and VEGFR-2 mRNA expression. CONCLUSION XTSJ decoction might inhibit angiogenesis in gastric cancer through IL-8-linked regulation of the VEGF pathway.
Collapse
Affiliation(s)
- Jun Shi
- Department of Traditional Chinese Medicine, Shanghai Changzheng Hospital, Second Military Medical University, No. 415 Fengyang Road, Huangpu District, Shanghai 200003, China.
| | - Ye Lu
- Department of Traditional Chinese Medicine, Shanghai Changzheng Hospital, Second Military Medical University, No. 415 Fengyang Road, Huangpu District, Shanghai 200003, China
| | - Pinkang Wei
- Department of Traditional Chinese Medicine, Shanghai Changzheng Hospital, Second Military Medical University, No. 415 Fengyang Road, Huangpu District, Shanghai 200003, China
| |
Collapse
|
19
|
Shi J, Wei PK. Interleukin-8: A potent promoter of angiogenesis in gastric cancer. Oncol Lett 2015; 11:1043-1050. [PMID: 26893688 DOI: 10.3892/ol.2015.4035] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 11/30/2015] [Indexed: 12/21/2022] Open
Abstract
Angiogenesis is a critical process in the development of tumor malignancy and occurs at various stages of tumor progression. Interleukin-8 (IL-8) is a pro-angiogenic factor produced by tumor-infiltrating macrophages that has been revealed to facilitate the development of angiogenesis in various cancers. However, whether IL-8 activates angiogenesis in gastric cancer remains unclear. The present study investigated the effect of IL-8 on the migration and canalization capacities of human umbilical vein endothelial cells (HUVECs). In addition, the protein and messenger RNA (mRNA) expression of selected angiogenesis markers, consisting of vascular endothelial growth factor (VEGF)-A, VEGF receptor (VEGFR)-1 and VEGFR-2, were assessed in the HUVECs. The HUVECs were co-cultured with human gastric cancer SGC7901 cells and exposed to various concentrations of IL-8 (0, 0.2, 0.5, 0.8 and 1.0 ng/ml). The migration and canalization abilities of the cells were detected by Transwell chamber and tube formation assays. Protein expression was detected using immunofluorescence and western blot analysis, and mRNA levels were assessed using reverse transcription quantitative polymerase chain reaction. The protein and mRNA levels of VEGF-A, VEGFR-1 and VEGFR-2 were measured in HUVECs cultured for 24 h. IL-8 at concentrations of 0.5, 0.8 and 1.0 ng/ml significantly promoted HUVEC cell migration (P=0.005, P=0.001 and P<0.001, respectively) and tube formation (P=0.039, P=0.003 and P<0.001, respectively). IL-8 at concentrations of 0.2, 0.5, 0.8 and 1.0 ng/ml significantly elevated the protein levels of VEGF-A (P<0.001) and VEGFR-2 (P=0.034, P<0.001, P<0.001 and P<0.001, respectively). IL-8 at concentrations of 0.8 and 1.0 ng/ml significantly elevated the protein levels of VEGF-1 (P=0.037 and P=0.002, respectively). Similarly, IL-8 at concentrations of 0.5, 0.8 and 1.0 ng/ml significantly upregulated the mRNA levels of VEGF-A (P=0.046, P=0.001 and P<0.001, respectively) and VEGFR-1 (P=0.042, P<0.001 and P<0.001, respectively). IL-8 at concentrations of 0.2, 0.5, 0.8 and 1.0 ng/ml significantly upregulated the mRNA levels of VEGFR-2 (P=0.003, P=0.005, P<0.001 and P<0.001, respectively). In conclusion, IL-8 may be a potent promoter of angiogenesis in gastric cancer.
Collapse
Affiliation(s)
- Jun Shi
- Department of Traditional Chinese Medicine, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai 200003, P.R. China
| | - Pin-Kang Wei
- Department of Traditional Chinese Medicine, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
20
|
Lugowska I, Kowalska M, Fuksiewicz M, Kotowicz B, Mierzejewska E, Koseła-Paterczyk H, Szamotulska K, Rutkowski P. Serum markers in early-stage and locally advanced melanoma. Tumour Biol 2015; 36:8277-85. [PMID: 26002577 PMCID: PMC4672018 DOI: 10.1007/s13277-015-3564-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/13/2015] [Indexed: 11/16/2022] Open
Abstract
The identification of prognostic factors in cutaneous melanoma allows choosing the most effective treatment, especially in group of patients with locoregional disease. Markers related to carcinogenesis and angiogenesis in particular have effect on the course of the disease. The aim of this study was to evaluate clinical utility of vascular endothelial growth factor (VEGF), matrix metalloproteinase 2 (MMP-2), MMP-9, tissue inhibitors of metalloproteinase 1 (TIMP-1), and YKL-40 in serum of melanoma patients at pathological stages I-III. We included 148 adult patients with melanoma. The median follow-up was 40 months. Disease recurrence was observed in 43 patients; 3-year disease-free survival (DFS) rate was 71.7%; 35 patients died; and the 3-year overall survival (OS) rate was 85%. Concentrations of VEGF, MMP-2, MMP-9, TIMP-1, and YKL-40 were measured by ELISA kits. VEGF, MMP-9, TIMP-1, and YKL-40 were significantly higher in group of patients than in controls. Increased concentrations of TIMP-1 were related to patient survival, which in the group of lower and increased TIMP-1, disease-free survival amounted to 81 vs. 61% (p = 0.014) and overall survival -88 vs. 82% (p = 0.050), respectively. An increased concentration of YKL-40 was observed in 59% of patients with ulceration and in 26% of patients without ulceration (p = 0.012). We have found a clinically significant correlation between YKL-40 and MMP-9 (rho = 0.363; p = 0.004) as well as YKL-40 and VEGF (rho = 0.306; p = 0.018). In melanoma patients at stages I-III, the high concentrations of TIMP-1 in serum predicted adverse prognosis. YKL-40 was associated with ulceration of primary tumor, which is a very important prognostic factor.
Collapse
Affiliation(s)
- Iwona Lugowska
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska Curie Memorial Cancer Centre and Institute of Oncology, K.W. Roentgen Street 5, Warsaw, Poland.
- Department of Epidemiology, Institute of Mother and Child, M. Kasprzak Street 17a, Warsaw, Poland.
| | - Maria Kowalska
- Department of Pathology and Laboratory Diagnostics, Laboratory of Tumor Markers, Maria Sklodowska Curie Memorial Cancer Centre and Institute of Oncology, K.W. Roentgen Street 5, Warsaw, Poland
| | - Małgorzata Fuksiewicz
- Department of Pathology and Laboratory Diagnostics, Laboratory of Tumor Markers, Maria Sklodowska Curie Memorial Cancer Centre and Institute of Oncology, K.W. Roentgen Street 5, Warsaw, Poland
| | - Beata Kotowicz
- Department of Pathology and Laboratory Diagnostics, Laboratory of Tumor Markers, Maria Sklodowska Curie Memorial Cancer Centre and Institute of Oncology, K.W. Roentgen Street 5, Warsaw, Poland
| | - Ewa Mierzejewska
- Department of Epidemiology, Institute of Mother and Child, M. Kasprzak Street 17a, Warsaw, Poland
| | - Hanna Koseła-Paterczyk
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska Curie Memorial Cancer Centre and Institute of Oncology, K.W. Roentgen Street 5, Warsaw, Poland
| | - Katarzyna Szamotulska
- Department of Epidemiology, Institute of Mother and Child, M. Kasprzak Street 17a, Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska Curie Memorial Cancer Centre and Institute of Oncology, K.W. Roentgen Street 5, Warsaw, Poland
| |
Collapse
|
21
|
Ott PA, Hodi FS, Buchbinder EI. Inhibition of Immune Checkpoints and Vascular Endothelial Growth Factor as Combination Therapy for Metastatic Melanoma: An Overview of Rationale, Preclinical Evidence, and Initial Clinical Data. Front Oncol 2015; 5:202. [PMID: 26442214 PMCID: PMC4585112 DOI: 10.3389/fonc.2015.00202] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/31/2015] [Indexed: 01/29/2023] Open
Abstract
The role of angiogenesis as a mediator of immune regulation in the tumor microenvironment has recently come into focus. Furthermore, emerging evidence indicates that immunotherapy can lead to immune-mediated vasculopathy in the tumor, suggesting that the tumor vasculature may be an important interface between the tumor-directed immune response and the cancer itself. The advent of immune checkpoint inhibition as an effective immunotherapeutic strategy for many cancers has led to a better understanding of this interface. While the inhibition of angiogenesis through targeting of vascular endothelial growth factor (VEGF) has been used successfully for the treatment of cancer for many years, the mechanisms of its anti-tumor activity remain poorly understood. Initial studies of the complex relationship between angiogenesis, VEGF signaling and the immune system suggest that the combination of immune checkpoint blockade with angiogenesis inhibition has potential. While the majority of this work has been performed in metastatic melanoma, immunotherapy is rapidly showing promise in a broad range of malignancies and efforts to enhance immunotherapy will broadly impact the future of oncology. Here, we review the preclinical rationale and clinical investigations of combined angiogenesis inhibition and immunotherapy/immune checkpoint inhibition as a potentially promising combinatorial approach for cancer treatment.
Collapse
Affiliation(s)
- Patrick A Ott
- Department of Medical Oncology, Melanoma Disease Center, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Harvard Medical School , Boston, MA , USA ; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA
| | - F Stephen Hodi
- Department of Medical Oncology, Melanoma Disease Center, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Harvard Medical School , Boston, MA , USA ; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA
| | - Elizabeth I Buchbinder
- Department of Medical Oncology, Melanoma Disease Center, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Harvard Medical School , Boston, MA , USA ; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
22
|
Yuan J, Zhou J, Dong Z, Tandon S, Kuk D, Panageas KS, Wong P, Wu X, Naidoo J, Page DB, Wolchok JD, Hodi FS. Pretreatment serum VEGF is associated with clinical response and overall survival in advanced melanoma patients treated with ipilimumab. Cancer Immunol Res 2015; 2:127-32. [PMID: 24778276 DOI: 10.1158/2326-6066.cir-13-0163] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ipilimumab, an antibody that blocks CTL antigen 4 (CTLA-4), improves overall survival (OS) for patients with metastatic melanoma. Given its role in angiogenesis and immune evasion, serum VEGF levels were evaluated for association with clinical benefit in ipilimumab-treated patients. Sera were collected from 176 patients treated at 3 (n = 98) or 10 mg/kg (n = 68). The VEGF levels before treatment and at induction completion (week 12) were analyzed using the Meso Scale Discovery kit. The association of the levels of VEGF with clinical responses and OS were assessed using the Fisher exact and Kaplan-Meier log-rank tests. VEGF as a continuous variable was associated with OS (P = 0.002). Using 43 pg/mL as the cutoff pretreatment VEGF value defined by maximally selected log-rank statistics, pretreatment VEGF values correlated with clinical benefit at week 24 (P = 0.019; 159 patients evaluable). Pretreatment VEGF ≥ 43 pg/mL was associated with decreased OS (median OS 6.6 vs. 12.9 months, P = 0.006; 7.4 vs. 14.3 months, P = 0.037 for 3 mg/kg; and 6.2 vs. 10.9 months, P = 0.048 for 10 mg/kg). There was no correlation between VEGF changes and clinical outcome. Serum VEGF may be a predictive biomarker for ipilimumab treatment and is worthy of prospective investigation with various forms of immunologic checkpoint blockade.
Collapse
Affiliation(s)
- Jianda Yuan
- Authors' Affiliations Melanoma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Al-Ameen MA, Li J, Beer DG, Ghosh G. Sensitive, quantitative, and high-throughput detection of angiogenic markers using shape-coded hydrogel microparticles. Analyst 2015; 140:4530-9. [DOI: 10.1039/c5an00358j] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Demonstration of the application of shape coded hydrogel microparticles for multiplexed detection of angiogenic molecules. Utilization of single fluorophore eliminates the spectral overlap associated with microparticle based multiplexed analysis.
Collapse
Affiliation(s)
- Mohammad Ali Al-Ameen
- Bioengineering Program
- Department of Mechanical Engineering
- University of Michigan
- Dearborn
- USA
| | - Ji Li
- Bioengineering Program
- Department of Mechanical Engineering
- University of Michigan
- Dearborn
- USA
| | - David G. Beer
- Thoracic Surgery
- Department of Surgery
- University of Michigan Comprehensive Cancer Center
- Ann Arbor MI 48109
- USA
| | - Gargi Ghosh
- Bioengineering Program
- Department of Mechanical Engineering
- University of Michigan
- Dearborn
- USA
| |
Collapse
|
24
|
Kendra KL, Plummer R, Salgia R, O'Brien MER, Paul EM, Suttle AB, Compton N, Xu CF, Ottesen LH, Villalona-Calero MA. A multicenter phase I study of pazopanib in combination with paclitaxel in first-line treatment of patients with advanced solid tumors. Mol Cancer Ther 2014; 14:461-9. [PMID: 25504632 DOI: 10.1158/1535-7163.mct-14-0431] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study was designed to evaluate the safety, pharmacokinetics, and clinical activity of pazopanib combined with paclitaxel to determine the recommended phase II dose in the first-line setting in patients with advanced solid tumors. Patients were enrolled in a 3+3 dose-escalation design to determine the maximum tolerated regimen (MTR) of once daily pazopanib plus paclitaxel administered every 3 weeks at four dose levels (DL1-4). Safety, pharmacokinetics, pharmacogenetics, and disease assessments were performed. Twenty-eight patients received treatment. One patient at DL1 had dose-limiting toxicity (DLT) of elevated hepatic enzymes. After pazopanib discontinuation, liver enzyme concentrations remained high until a concurrent medication, simvastatin, was discontinued. This patient had the defective CYP2C8*3*3 genotype. At DL2, 1 patient had DLT of elevated hepatic enzymes with rash and 1 patient had DLT of rash. The MTR was paclitaxel 150 mg/m(2) plus pazopanib 800 mg. The most common toxicities were alopecia, fatigue, hypertension, nausea, diarrhea, dysgeusia, neutropenia, myalgia, hair color changes, and peripheral neuropathy. Coadministration of pazopanib and paclitaxel resulted in a 38% increase in systemic exposure to paclitaxel, relative to administration of paclitaxel alone, at the MTR. Of the 28 patients treated with the combination, 10 achieved a partial response and 10 achieved stable disease of ≥12 weeks. Pazopanib 800 mg daily plus paclitaxel 150 mg/m(2) every 3 weeks was the recommended phase II dose, with a manageable safety profile, and with clinical activity in both melanoma and non-small cell lung cancer that suggest further evaluation of this combination is warranted.
Collapse
Affiliation(s)
- Kari L Kendra
- Division of Medical Oncology and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States.
| | - Ruth Plummer
- Sir Bobby Robson Cancer Trials Research Centre, Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | | | - Mary E R O'Brien
- Department of Medicine, Royal Marsden NHS Foundation Trust, Sutton, Surrey, United Kingdom
| | - Elaine M Paul
- Oncology Research and Development, GlaxoSmithKline, Research Triangle Park, North Carolina
| | - A Benjamin Suttle
- Oncology Research and Development, GlaxoSmithKline, Research Triangle Park, North Carolina
| | - Natalie Compton
- Oncology Research and Development, GlaxoSmithKline, Uxbridge, United Kingdom
| | - Chun-Fang Xu
- Oncology Research and Development, GlaxoSmithKline, Uxbridge, United Kingdom
| | - Lone H Ottesen
- Oncology Research and Development, GlaxoSmithKline, Uxbridge, United Kingdom
| | - Miguel A Villalona-Calero
- Division of Medical Oncology and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
25
|
Expression of angiogenesis regulatory proteins and epithelial-mesenchymal transition factors in platelets of the breast cancer patients. ScientificWorldJournal 2014; 2014:878209. [PMID: 25379550 PMCID: PMC4212629 DOI: 10.1155/2014/878209] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 08/14/2014] [Indexed: 12/21/2022] Open
Abstract
Platelets play a role in tumor angiogenesis and growth and are the main transporters of several angiogenesis regulators. Here, we aimed to determine the levels of angiogenesis regulators and epithelial-mesenchymal transition factors sequestered by circulating platelets in breast cancer patients and age-matched healthy controls. Platelet pellets (PP) and platelet-poor plasma (PPP) were collected by routine protocols. Vascular endothelial growth factor (VEGF), platelet-derived growth factor BB (PDGF-BB), thrombospondin-1 (TSP-1), platelet factor 4 (PF4), and transforming growth factor-β1 (TGF-β1) were measured by enzyme-linked immunosorbent assay. Angiogenesis-associated expression of VEGF (2.1 pg/106 platelets versus 0.9 pg/106 platelets, P < 0.001), PF4 (21.2 ng/106 platelets versus 10.2 ng/106 platelets, P < 0.001), PDGF-BB (42.9 pg/106 platelets versus 19.1 pg/106 platelets, P < 0.001), and TGF-β1 (15.3 ng/106 platelets versus 4.3 ng/106 platelets, P < 0.001) differed in the PP samples of cancer and control subjects. In addition, protein concentrations were associated with clinical characteristics (P < 0.05). Circulating platelets in breast cancer sequester higher levels of PF4, VEGF, PDGF-BB, and TGF-β1, suggesting a possible target for early diagnosis. VEGF, PDGF, and TGF-β1 concentrations in platelets may be associated with prognosis.
Collapse
|
26
|
Pastushenko I, Vermeulen PB, Van den Eynden GG, Rutten A, Carapeto FJ, Dirix LY, Van Laere S. Mechanisms of tumour vascularization in cutaneous malignant melanoma: clinical implications. Br J Dermatol 2014; 171:220-33. [PMID: 24641095 DOI: 10.1111/bjd.12973] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2014] [Indexed: 01/02/2023]
Abstract
Malignant melanoma represents < 10% of all skin cancers but is responsible for the majority of skin-cancer-related deaths. Metastatic melanoma has historically been considered as one of the most therapeutically challenging malignancies. Fortunately, for the first time after decades of basic research and clinical investigation, new drugs have produced major clinical responses. Angiogenesis has been considered an important target for cancer treatment. Initial efforts have focused primarily on targeting endothelial and tumour-related vascular endothelial growth factor signalling. Here, we review different mechanisms of tumour vascularization described in melanoma and discuss the potential clinical implications.
Collapse
Affiliation(s)
- I Pastushenko
- Department of Dermatology, Hospital Clínico Universitario 'Lozano Blesa', Zaragoza, 50009, Spain
| | | | | | | | | | | | | |
Collapse
|
27
|
Carvajal RD, Wong MK, Thompson JA, Gordon MS, Lewis KD, Pavlick AC, Wolchok JD, Rojas PB, Schwartz JD, Bedikian AY. A phase 2 randomised study of ramucirumab (IMC-1121B) with or without dacarbazine in patients with metastatic melanoma. Eur J Cancer 2014; 50:2099-107. [PMID: 24930625 PMCID: PMC5702465 DOI: 10.1016/j.ejca.2014.03.289] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 01/27/2014] [Accepted: 03/26/2014] [Indexed: 01/02/2023]
Abstract
BACKGROUND To evaluate the efficacy and safety of ramucirumab (IMC-1121B; LY3009806), a fully human monoclonal antibody targeting the vascular endothelial growth factor receptor-2, alone and in combination with dacarbazine in chemotherapy-naïve patients with metastatic melanoma (MM). METHODS Eligible patients received ramucirumab (10mg/kg) + dacarbazine (1000 mg/m(2)) (Arm A) or ramucirumab only (10mg/kg) (Arm B) every 3 weeks. The primary end-point was progression-free survival (PFS); secondary end-points included overall survival (OS), overall response and safety. FINDINGS Of 106 randomised patients, 102 received study treatment (Arm A, N=52; Arm B, N=50). Baseline characteristics were similar in both arms. Median PFS was 2.6 months (Arm A) and 1.7 months (Arm B); median 6-month PFS rates were 30.7% and 17.9% and 12-month PFS rates were 23.7% and 15.6%, respectively. In Arm A, 9 (17.3%) patients had partial response (PR) and 19 (36.5%), stable disease (SD); PR and SD in Arm B were 2 (4.0%) and 21 (42.0%), respectively. Median OS was 8.7 months in Arm A and 11.1 months in Arm B. Patients in both arms tolerated the treatment with limited Grade 3/4 toxicities. INTERPRETATION Ramucirumab alone or in combination with dacarbazine was associated with an acceptable safety profile in patients with MM. Although the study was not powered for comparison between treatment arms, PFS appeared greater with combination therapy. Sustained disease control was observed on both study arm.
Collapse
Affiliation(s)
| | - Michael K Wong
- USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | | | | | | | | | - Jedd D Wolchok
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Patrick B Rojas
- ImClone Systems LLC, a wholly-owned subsidiary of Eli Lilly and Company, Bridgewater, NJ, USA
| | - Jonathan D Schwartz
- ImClone Systems LLC, a wholly-owned subsidiary of Eli Lilly and Company, Bridgewater, NJ, USA
| | | |
Collapse
|
28
|
Tas F, Karabulut S, Yasasever CT, Duranyildiz D. Serum transforming growth factor-beta 1 (TGF-β1) levels have diagnostic, predictive, and possible prognostic roles in patients with melanoma. Tumour Biol 2014; 35:7233-7. [PMID: 24771267 DOI: 10.1007/s13277-014-1984-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 04/17/2014] [Indexed: 10/25/2022] Open
Abstract
Transforming growth factor-beta 1 (TGF-β1) plays an important role in the pathogenesis of multiple malignancies, and its expression also strongly affects the outcomes of cancer patients. The objective of this study was to determine the clinical significance of the serum levels of TGF-β1 in melanoma patients. A total of 60 patients with a pathologically confirmed diagnosis of melanoma were enrolled into this study. Serum TGF-β1 concentrations were determined by the solid-phase sandwich ELISA method. Thirty age- and sex-matched healthy controls were included in the analysis. The median age at diagnosis was 53.5 years (range 16 to 88 years). The baseline serum TGF-β1 levels of the melanoma patients were significantly higher than those in the control group (median values 171.85 vs. 19.95 pg/mL, respectively; p < 0.001). The known clinical variables including age of patient, gender, site of lesion, histology, stage of disease, and serum LDH levels were not found to be correlated with serum TGF-β1 concentrations (p > 0.05). However, the chemotherapy-responsive patients had higher serum TGF-β1 levels compared with chemotherapy-unresponsive ones (p = 0.05). Additionally, serum TGF-β1 concentration was a trend to have a prognostic role on survival (p = 0.07). Patients with elevated serum TGF-β1 concentrations had close to significantly favorable overall survival compared to those with lower levels (median 30.1 vs. 20.9 months, respectively). In conclusion, serum levels of TGF-β1 have diagnostic, predictive, and possible prognostic roles in melanoma patients.
Collapse
Affiliation(s)
- Faruk Tas
- Institute of Oncology, University of Istanbul, 34390, Capa, Istanbul, Turkey,
| | | | | | | |
Collapse
|
29
|
Kaufman KL, Mactier S, Armstrong NJ, Mallawaaratchy D, Byrne SN, Haydu LE, Jakrot V, Thompson JF, Mann GJ, Scolyer RA, Christopherson RI. Surface antigen profiles of leukocytes and melanoma cells in lymph node metastases are associated with survival in AJCC stage III melanoma patients. Clin Exp Metastasis 2014; 31:407-21. [PMID: 24435119 PMCID: PMC3973954 DOI: 10.1007/s10585-014-9636-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 01/09/2014] [Indexed: 12/14/2022]
Abstract
There is an urgent need to identify more accurate prognostic biomarkers in melanoma patients, particularly in those with metastatic disease. This study aimed to identify melanoma and leukocyte surface antigens predictive of survival in a prospective series of AJCC stage IIIb/c melanoma patients (n = 29). Live cell suspensions were prepared from melanoma metastases within lymph nodes (LN). The suspensions were immuno-magnetically separated into CD45+ (leukocyte) and CD45− (non-hematopoietic, enriched melanoma cell) fractions. Surface antigens on CD45− and CD45+ cell populations were profiled using DotScan™ microarrays (Medsaic Pty. Ltd.) and showed differential abundance levels for 52 and 78 antigens respectively. Associations of the surface profiles with clinicopathologic and outcome data (median follow-up 35.4 months post LN resection) were sought using univariate (log-rank test) and multivariate (Wald’s test; modelled with patient’s age, gender and AJCC staging at LN recurrence) survival models. CD9 (p = 0.036), CD39 (p = 0.004) and CD55 (p = 0.005) on CD45+ leukocytes were independently associated with distant metastasis-free survival using multivariate analysis. Leukocytes with high CD39 levels were also significantly associated with increased overall survival (OS) in multivariate analysis (p = 0.016). LNs containing leukocytes expressing CD11b (p = 0.025), CD49d (p = 0.043) and CD79b (p = 0.044) were associated with reduced OS on univariate analysis. For enriched melanoma cells (CD45− cell populations), 11 surface antigens were significantly correlated with the disease-free interval (DFI) between diagnosis of culprit primary melanoma and LN metastasis resection. Nine antigens on CD45+ leukocytes also correlated with DFI. Following validation in independent datasets, surface markers identified here should enable more accurate determination of prognosis in stage III melanoma patients and provide better risk stratification of patients entering clinical trials.
Collapse
Affiliation(s)
- Kimberley L Kaufman
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, 2006, Australia.
| | - Swetlana Mactier
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, 2006, Australia
| | - Nicola J Armstrong
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,School of Mathematics and Statistics and Prince of Wales Clinical School, University of New South Wales, Kensington, NSW, 2052, Australia
| | | | - Scott N Byrne
- Discipline of Infectious Diseases and Immunology Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia.,Discipline of Dermatology, Bosch Institute, Faculty of Medicine, Sydney Medical School, Sydney, NSW, 2006, Australia
| | - Lauren E Haydu
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia.,Discipline of Surgery, Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Valerie Jakrot
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia
| | - John F Thompson
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia
| | - Graham J Mann
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia.,Westmead Institute of Cancer Research, The University of Sydney at Westmead Millennium Institute, Westmead, NSW, 2145, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia.,Discipline of Pathology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | | |
Collapse
|
30
|
|
31
|
Zbytek B, Carlson JA, Granese J, Ross J, Mihm MC, Slominski A. Current concepts of metastasis in melanoma. ACTA ACUST UNITED AC 2014; 3:569-585. [PMID: 19649148 DOI: 10.1586/17469872.3.5.569] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The main cause of death in melanoma patients is widespread metastases. Staging of melanoma is based on the primary tumor thickness, ulceration, lymph node and distant metastases. Metastases develop in regional lymph nodes, as satellite or in-transit lesions, or in distant organs. Lymph flow and chemotaxis is responsible for the homing of melanoma cells to different sites. Standard pathologic evaluation of sentinel lymph nodes fails to find occult melanoma in a significant proportion of cases. Detection of small numbers of malignant melanoma cells in these and other sites, such as adjacent to the primary site, bone marrow or the systemic circulation, may be enhanced by immunohistochemistry, reverse transcription PCR, evaluation of lymphatic vessel invasion and proteomics. In the organs to which melanoma cells metastasize, extravasation of melanoma cells is regulated by adhesion molecules, matrix metalloproteases, chemokines and growth factors. Melanoma cells may travel along external vessel lattices. After settling in the metastatic sites, melanoma cells develop mechanisms that protect them against the attack of the immune system. It is thought that one of the reasons why melanoma cells are especially resistant to killing is the fact that melanocytes (cells from which melanoma cells derive) are resistant to such noxious factors as ultraviolet light and reactive oxygen species. Targeted melanoma therapies are, so far, largely unsuccessful, and new ones, such as adjuvant inhibition of melanogenesis, are under development.
Collapse
Affiliation(s)
- Blazej Zbytek
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, 930 Madison Avenue, Memphis, TN 38163, USA, Tel.: +1 901 448 6300, ,
| | | | | | | | | | | |
Collapse
|
32
|
|
33
|
Mansfield AS, Markovic SN. Inhibition of Angiogenesis for the Treatment of Metastatic Melanoma. Curr Oncol Rep 2013; 15:492-9. [DOI: 10.1007/s11912-013-0334-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
34
|
A prospective study of 67 serum immune and inflammation markers and risk of non-Hodgkin lymphoma. Blood 2013; 122:951-7. [PMID: 23814017 DOI: 10.1182/blood-2013-01-481077] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Although severe immune dysregulation is an established risk factor for non-Hodgkin lymphoma (NHL), the importance of subclinical immunologic effects is unclear. We compared baseline serum levels of 67 immune and inflammation markers between 301 patients with NHL diagnosed 5+ years after blood collection and 301 control patients within the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. We observed associations with NHL for elevated B-cell-attracting chemokine 1 (BCA-1; fourth quartile vs first: odds ratio [OR], 2.7; 95% confidence interval [CI], 1.7-4.2; Ptrend = 1.0 × 10(-6)), soluble tumor necrosis factor receptor 2 (sTNFR2; OR, 3.4; 95% CI, 2.0-5.8; Ptrend = 1.1 × 10(-6)), and soluble vascular endothelial growth factor receptor 2 (sVEGFR2; OR, 2.3; 95% CI, 1.4-3.9; Ptrend = .0005) that remained significant after Bonferroni correction, simultaneous model adjustment, and restriction to cases diagnosed 8 to 13 years after blood collection. Associations with other markers were observed, although none remained associated with NHL after adjustment for BCA-1, sTNFR2, and sVEGFR2. Our findings suggest that circulating BCA-1, sTNFR2, and sVEGFR2 are associated with NHL risk well in advance of diagnosis. Additional research is needed to replicate these findings and elucidate the underlying biologic mechanisms.
Collapse
|
35
|
Circulating levels of VEGFR-1 and VEGFR-2 in patients with metastatic melanoma treated with chemoimmunotherapy alone or combined with bevacizumab. Melanoma Res 2012; 21:431-7. [PMID: 21730877 DOI: 10.1097/cmr.0b013e32834941d3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
There are no identified biomarkers that could predict response to antiangiogenic or traditional chemoimmunotherapy in metastatic melanoma. We hypothesized that soluble angiogenic factor receptors might help us to identify patients responsive to treatment. A series of 48 patients with stage IV melanoma participating in two phase II clinical trials were included. The trials included treatment with carboplatin, vinorelbine, and subcutaneous interleukin-2 (n=22) or treatment with bevacizumab, dacarbazine, and low-dose interferon-α2a (n=26).Serum samples were prospectively collected and soluble vascular endothelial growth factor receptor 1 (s-VEGFR-1) and 2 (s-VEGFR-2) were measured before starting the trial treatment and during response evaluation.There was a trend toward longer overall survival among patients with higher-than-median serum VEGFR-1 levels (21.3 months) compared with 12.3 months in patients with low pretreatment s-VEGFR-1 levels (P=0.146). Pretreatment s-VEGFR-2 levels did not correlate to survival. Serum VEGFR-2 levels decreased during therapy in 44% of the patients and increased in 56% of the patients. VEGFR-2 increased in 78% (14 of 18) of the patients who progressed during therapy (P=0.017). VEGFR-2 decrease was associated with clinical benefit in 65% of the patients (11 of 17) and with progression in only four patients (P=0.016).High pretreatment levels of s-VEGFR-1 are associated with improved prognosis among patients with metastatic melanoma independently on therapy, whereas increased VEGFR-2 levels during therapy are associated with disease progression. These markers might be useful in selecting patients responsive to antiangiogenic therapy.
Collapse
|
36
|
Tarhini AA, Frankel P, Margolin KA, Christensen S, Ruel C, Shipe-Spotloe J, Gandara DR, Chen A, Kirkwood JM. Aflibercept (VEGF Trap) in inoperable stage III or stage iv melanoma of cutaneous or uveal origin. Clin Cancer Res 2011; 17:6574-81. [PMID: 21880788 DOI: 10.1158/1078-0432.ccr-11-1463] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Aflibercept is a soluble decoy VEGF receptor and angiogenesis inhibitor with potent preclinical antitumor activity in melanoma. We conducted a multicenter phase II study in patients with inoperable stage III or IV melanoma and no prior chemotherapy. EXPERIMENTAL DESIGN A two-stage design was adopted to evaluate 4-month progression-free survival rate (PFSR) and response rate. Aflibercept was given at 4 mg/kg intravenously every 2 weeks. Response was assessed every 8 weeks. First-stage accrual of 21 patients was specified and with an adequate 4-month PFSR accrual continued to a total of 41. RESULTS Forty-one patients of ages 23 to 84 (median = 57) were enrolled. Thirty-nine had American Joint Committee on Cancer stage IV (5 M1a, 7 M1b, and 27 M1c) and 2 had inoperable stage IIIC (N3). Eastern Cooperative Oncology Group (ECOG) performance status was 0 (27 patients) or 1 (14 patients). Ten patients had primary uveal melanoma, 28 cutaneous, and 3 had unknown primaries. A median of 7 cycles were initiated (range: 1-56). Grade 3 and 4 toxicities included hypertension in 9 patients (22%) and proteinuria in 6 (15%). Among 40 patients evaluable for efficacy (those who initiated aflibercept), 3 (7.5%) had a confirmed partial response and 20 had progression-free survival of 4 months or above. The predicted 1-year survival rate derived from the Korn meta-analysis model is 36% (N = 39), whereas we observed a corresponding 56.4% survival rate at 1 year (95% CI, 43-74, P < 0.005). Median overall survival in this trial is 16.3 months (95% CI, 9.2 to not reached). We observed a significant association between severity of hypertension following aflibercept and survival improvement. CONCLUSIONS Aflibercept showed promising activity in patients with metastatic melanoma of cutaneous or uveal origin. Further evaluation of aflibercept as a single agent and in combination is warranted.
Collapse
|
37
|
Rykala J, Przybylowska K, Majsterek I, Pasz-Walczak G, Sygut A, Dziki A, Kruk-Jeromin J. Angiogenesis markers quantification in breast cancer and their correlation with clinicopathological prognostic variables. Pathol Oncol Res 2011; 17:809-17. [PMID: 21560015 PMCID: PMC3185224 DOI: 10.1007/s12253-011-9387-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 03/09/2011] [Indexed: 12/29/2022]
Abstract
Tumoural angiogenesis is essential for the growth and spread of breast cancer cells. Therefore the aim of this study was to assess the diagnostic performance of angiogenesis markers in tumours and there reflecting levels in serum of breast cancer patients. Angiogenin, Ang2, fibroblast growth factor basic, intercellular adhesion molecule (ICAM)-1, keratinocyte growth factor (KGF), platelet-derived growth factor-BB, and VEGF-A were measured using a FASTQuant angiogenic growth factor multiplex protein assay. We observed that breast cancer tumours exhibited high levels of PDGF-BB, bFGF and VEGF, and extremely high levels of TIMP-1 and Ang-2, whereas in serum we found significantly higher levels of Ang-2, PDGF-BB, bFGF, ICAM-1 and VEGF in patients with breast cancer compared to the benign breast diseases patients. Moreover, some of these angiogenesis markers evaluated in tumour and serum of breast cancer patients exhibited association with standard clinical parameters, ER status as well as MVD of tumours. Angiogenesis markers play important roles in tumour growth, invasion and metastasis. Our results suggest that analysis of angiogenesis markers in tumour and serum of breast cancer patients using multiplex protein assay can improve diagnosis and prognosis in this diseases.
Collapse
Affiliation(s)
- Jan Rykala
- Department of Plastic, Reconstructive and Aesthetic Surgery, Barlicki Hospital, Medical University of Lodz, Kopcinskiego 22, 90-153, Lodz, Poland.
| | | | | | | | | | | | | |
Collapse
|
38
|
Nagengast WB, Hooge MNLD, van Straten EME, Kruijff S, Brouwers AH, den Dunnen WFA, de Jong JR, Hollema H, Dierckx RA, Mulder NH, de Vries EGE, Hoekstra HJ, Hospers GAP. VEGF-SPECT with ¹¹¹In-bevacizumab in stage III/IV melanoma patients. Eur J Cancer 2011; 47:1595-602. [PMID: 21429739 DOI: 10.1016/j.ejca.2011.02.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 02/14/2011] [Indexed: 11/25/2022]
Abstract
PURPOSE A feasibility study was performed to investigate the presence of VEGF in melanoma lesions by VEGF-SPECT with (111)In-bevacizumab. In addition the effect of a single therapeutic bevacizumab dose on (111)In-bevacizumab uptake was compared with VEGF levels in resected melanoma lesions. PATIENTS AND METHODS Eligible were patients with stage III/IV melanoma who presented with nodal recurrent disease. VEGF-SPECT was performed after administration of 100 Mbq (111)In-bevacizumab (8 mg) at days 0, 2, 4 and 7 post injection. Tumour visualisation and quantification were compared with CT and FDG-PET. On day 7 a single dose of 7.5mg/kg bevacizumab was administered intravenously. On day 21, a second tracer dose (111)In-bevacizumab was administered and scans were obtained on days 21, 25 and 28. Metastases were surgically resected within 2 weeks after the last VEGF-SPECT scan and immunohistological (IHC) VEGF tumour expression was compared with (111)In-bevacizumab tumour uptake. RESULTS Nine patients were included. FDG-PET and CT detected both in total 12 nodal lesions which were all visualised by VEGF-SPECT. At baseline, (111)In-bevacizumab tumour uptake varied 3-fold between and 1.6 ± 0.1-fold within patients. After a therapeutic dose of bevacizumab there was a 21 ± 4% reduction in (111)In-bevacizumab uptake. The (111)In-bevacizumab tumour uptake in the second series positively correlated with the VEGF-A expression in the resected tumour lesions. CONCLUSION VEGF-SPECT could visualise all known melanoma lesions. A single dose of bevacizumab slightly lowered (111)In-bevacizumab uptake. Future studies should elucidate the role of VEGF-SPECT in the selection of patients and the individual dosing of bevacizumab treatment.
Collapse
Affiliation(s)
- Wouter B Nagengast
- Department of Medical Oncology, University Medical Center Groningen, Hanzeplein 1, Postbus 30.001, 9700 RB Groningen, Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Bennuru S, Maldarelli G, Kumaraswami V, Klion AD, Nutman TB. Elevated levels of plasma angiogenic factors are associated with human lymphatic filarial infections. Am J Trop Med Hyg 2010; 83:884-90. [PMID: 20889885 DOI: 10.4269/ajtmh.2010.10-0039] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Lymphatic dilatation, dysfunction, and lymphangiogenesis are hallmarks of patent lymphatic filariasis, observed even in those with subclinical microfilaremia, through processes associated, in part, by vascular endothelial growth factors (VEGFs). A panel of pro-angiogenic factors was measured in the plasma of subjects from filaria-endemic regions using multiplexed immunological assays. Compared with endemic normal control subjects, those with both subclinical microfilaremia, and those with longstanding lymphedema had significantly elevated levels of VEGF-A, VEGF-C, VEGF-D, and angiopoietins (Ang-1/Ang-2), with only levels of basic fibroblast growth factor (bFGF) and placental growth factor (PlGF) being elevated only if lymphedema was evident. Furthermore, levels of these factors 1-year post-treatment with doxycycline were similar to pretreatment levels suggesting a minimal role, if any, for Wolbachia. Our data support the concept that filarial infection per se is associated with elevated levels of most of the known pro-angiogenic factors, with only a few being associated with the serious pathologic consequences associated with Wuchereria bancrofti infection.
Collapse
Affiliation(s)
- Sasisekhar Bennuru
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | |
Collapse
|
40
|
Del Vecchio M, Mortarini R, Canova S, Di Guardo L, Pimpinelli N, Sertoli MR, Bedognetti D, Queirolo P, Morosini P, Perrone T, Bajetta E, Anichini A. Bevacizumab plus fotemustine as first-line treatment in metastatic melanoma patients: clinical activity and modulation of angiogenesis and lymphangiogenesis factors. Clin Cancer Res 2010; 16:5862-72. [PMID: 21030496 DOI: 10.1158/1078-0432.ccr-10-2363] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To assess the clinical and biological activity of the association of bevacizumab and fotemustine as first-line treatment in advanced melanoma patients. EXPERIMENTAL DESIGN Previously untreated, metastatic melanoma patients (n = 20) received bevacizumab (at 15 mg/kg every 3 weeks) and fotemustine (100 mg/m² by intravenous administration on days 1, 8, and 15, repeated after 4 weeks) in a multicenter, single-arm, open-label, phase II study. Primary endpoint was the best overall response rate; other endpoints were toxicity, time to progression (TTP), and overall survival (OS). Serum cytokines, angiogenesis, and lymphangiogenesis factors were monitored by multiplex arrays and by in vitro angiogenesis assays. Effects of fotemustine on melanoma cells, in vitro, on vascular endothelial growth factor (VEGF)-C release and apoptosis were assessed by ELISA and flow cytometry, respectively. RESULTS One complete response, 2 partial responses (PR), and 10 patients with stable disease were observed. TTP and OS were 8.3 and 20.5 months, respectively. Fourteen patients experienced adverse events of toxicity grade 3-4. Serum VEGF-A levels in evaluated patients (n = 15) and overall serum proangiogenic activity were significantly inhibited. A significant reduction in VEGF-C levels was found in several post-versus pretherapy serum samples. In vitro, fotemustine inhibited VEGF-C release by melanoma cells without inducing significant cell death. Serum levels of interleukin (IL)-10 and IL-12p70 showed the highest levels in sera of PR patients, compared with patients with stable or progressive disease whereas IL-23 showed the opposite pattern. CONCLUSIONS The combination of bevacizumab plus fotemustine has clinical activity in advanced melanoma and promotes systemic modulation of angiogenesis and lymphangiogenesis factors.
Collapse
Affiliation(s)
- Michele Del Vecchio
- Unit of Medical Oncology 2, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
A phase II trial of bevacizumab with dacarbazine and daily low-dose interferon-alpha2a as first line treatment in metastatic melanoma. Melanoma Res 2010; 20:318-25. [PMID: 20375744 DOI: 10.1097/cmr.0b013e3283390365] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Metastatic melanomas are hypervascular tumours with poor prognosis. We hypothesized that treatment of metastatic melanoma with a combination of bevacizumab, a monoclonal antibody against vascular endothelial growth factor, dacarbazine (DTIC) and low-dose interferon alpha-2a (IFN-alpha2a) might lead to a synergistic inhibition of angiogenesis and regression of tumours. Patients with metastatic melanoma were treated with bevacizumab (5 mg/kg every 2 weeks), DTIC (200 mg/m days 1-5 every 4 weeks) and IFN-alpha2a (three MIU subcutaneously daily from day 15 onwards). Patients exhibiting response or stable disease after 6 months were treated with bevacizumab+/-IFN-alpha2a until disease progression. The primary study objectives were progression-free survival (PFS), overall survival and safety. Twenty-six patients were accrued. Response rate was 23% (two complete responses, four partial responses), and six patients showed stable disease. The median PFS for all patients was 2.3 months and for responders 8.1 months. The median overall survival for all patients was 11.5 months. Four life-threatening adverse events were seen: two pulmonary thromboembolisms, an intracerebral haemorrhage, and one grade 4 hypertension. One of the pulmonary emboli and the intracerebral haemorrhage were observed > or =3 months after the last bevacizumab-DTIC dose. Serum matrix metalloproteinase-9 and vascular endothelial growth factor levels changed during therapy. There was a trend towards favourable PFS among patients with only minimal or moderate change in these marker expression levels. The present regimen was active in this patient group but was also associated with remarkable vascular events.
Collapse
|
42
|
Chaiworapongsa T, Kusanovic JP, Savasan ZA, Mazaki-Tovi S, Kim SK, Vaisbuch E, Tarca AL, Mittal P, Ogge G, Madan I, Dong Z, Yeo L, Hassan SS, Romero R. Fetal death: a condition with a dissociation in the concentrations of soluble vascular endothelial growth factor receptor-2 between the maternal and fetal compartments. J Matern Fetal Neonatal Med 2010; 23:960-72. [PMID: 20158395 PMCID: PMC3427783 DOI: 10.3109/14767050903410664] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE An anti-angiogenic state has been implicated in the pathophysiology of preeclampsia, fetal growth restriction and fetal death. Vascular endothelial growth factor (VEGF), an indispensible angiogenic factor for embryonic and placental development exerts its angiogenic properties through the VEGF receptor (VEGFR)-2. A soluble form of this protein (sVEGFR-2) has been recently detected in maternal blood. The aim of this study was to determine if fetal death was associated with changes in the concentrations of sVEGFR-2 in maternal plasma and amniotic fluid. STUDY DESIGN Maternal plasma was obtained from patients with fetal death (n = 59) and normal pregnant women (n = 134). Amniotic fluid was collected from 36 patients with fetal death and the control group consisting of patients who had an amniocentesis and delivered at term (n = 160). Patients with fetal death were classified according to the clinical circumstances into the following groups: (1) unexplained; (2) preeclampsia and/or placental abruption; (3) chromosomal and/or congenital anomalies. Plasma and amniotic fluid concentrations of sVEGFR-2 were determined by ELISA. Non-parametric statistics and logistic regression analysis were applied. RESULTS (1) Patients with a fetal death had a significantly lower median plasma concentration of sVEGFR-2 than normal pregnant women (p < 0.001). The median plasma concentration of sVEGFR-2 in patients with unexplained fetal death and in those with preeclampsia/abruption, but not that of those with congenital anomalies, was lower than that of normal pregnant women (p = 0.006, p < 0.001 and p = 0.2, respectively); (2) the association between plasma sVEGFR-2 concentrations and preterm unexplained fetal death remained significant after adjusting for potential confounders (OR: 3.2; 95% CI: 1.4-7.3 per each quartile decrease in plasma sVEGFR-2 concentrations); (3) each subgroup of fetal death had a higher median amniotic fluid concentration of sVEGFR-2 than the control group (p < 0.001 for each); (4) the association between amniotic fluid sVEGFR-2 concentrations and preterm unexplained fetal death remained significant after adjusting for potential confounders (OR: 15.6; 95% CI: 1.5-164.2 per each quartile increase in amniotic fluid sVEGFR-2 concentrations); (5) among women with fetal death, there was no relationship between maternal plasma and amniotic fluid concentrations of sVEGFR-2 (Spearman Rho: 0.02; p = 0.9). CONCLUSION Pregnancies with a fetal death, at the time of diagnosis, are characterized by a decrease in the maternal plasma concentration of sVEGFR-2, but an increase in the amniotic fluid concentration of this protein. Although a decrease in sVEGFR-2 concentration in maternal circulation depends upon the clinical circumstances of fetal death, an increase in sVEGFR-2 concentration in amniotic fluid seems to be a common feature of fetal death. It remains to be determined if the perturbation in sVEGFR-2 concentrations in maternal and fetal compartments observed herein preceded the death of a fetus.
Collapse
Affiliation(s)
- Tinnakorn Chaiworapongsa
- Perinatology Research Branch, NICHD, NIH, DHHS, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Juan Pedro Kusanovic
- Perinatology Research Branch, NICHD, NIH, DHHS, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Zeynep Alpay Savasan
- Perinatology Research Branch, NICHD, NIH, DHHS, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Shali Mazaki-Tovi
- Perinatology Research Branch, NICHD, NIH, DHHS, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Sun Kwon Kim
- Perinatology Research Branch, NICHD, NIH, DHHS, Bethesda, MD and Detroit, MI, USA
| | - Edi Vaisbuch
- Perinatology Research Branch, NICHD, NIH, DHHS, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Adi L Tarca
- Perinatology Research Branch, NICHD, NIH, DHHS, Bethesda, MD and Detroit, MI, USA
- Department of Computer Science, Wayne State University, Detroit, MI
| | - Pooja Mittal
- Perinatology Research Branch, NICHD, NIH, DHHS, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Giovanna Ogge
- Perinatology Research Branch, NICHD, NIH, DHHS, Bethesda, MD and Detroit, MI, USA
| | - Ichchha Madan
- Perinatology Research Branch, NICHD, NIH, DHHS, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Zhong Dong
- Perinatology Research Branch, NICHD, NIH, DHHS, Bethesda, MD and Detroit, MI, USA
| | - Lami Yeo
- Perinatology Research Branch, NICHD, NIH, DHHS, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Sonia S Hassan
- Perinatology Research Branch, NICHD, NIH, DHHS, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, NICHD, NIH, DHHS, Bethesda, MD and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| |
Collapse
|
43
|
Berretta R, Moscato P. Cancer biomarker discovery: the entropic hallmark. PLoS One 2010; 5:e12262. [PMID: 20805891 PMCID: PMC2923618 DOI: 10.1371/journal.pone.0012262] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2009] [Accepted: 06/26/2010] [Indexed: 12/29/2022] Open
Abstract
Background It is a commonly accepted belief that cancer cells modify their transcriptional state during the progression of the disease. We propose that the progression of cancer cells towards malignant phenotypes can be efficiently tracked using high-throughput technologies that follow the gradual changes observed in the gene expression profiles by employing Shannon's mathematical theory of communication. Methods based on Information Theory can then quantify the divergence of cancer cells' transcriptional profiles from those of normally appearing cells of the originating tissues. The relevance of the proposed methods can be evaluated using microarray datasets available in the public domain but the method is in principle applicable to other high-throughput methods. Methodology/Principal Findings Using melanoma and prostate cancer datasets we illustrate how it is possible to employ Shannon Entropy and the Jensen-Shannon divergence to trace the transcriptional changes progression of the disease. We establish how the variations of these two measures correlate with established biomarkers of cancer progression. The Information Theory measures allow us to identify novel biomarkers for both progressive and relatively more sudden transcriptional changes leading to malignant phenotypes. At the same time, the methodology was able to validate a large number of genes and processes that seem to be implicated in the progression of melanoma and prostate cancer. Conclusions/Significance We thus present a quantitative guiding rule, a new unifying hallmark of cancer: the cancer cell's transcriptome changes lead to measurable observed transitions of Normalized Shannon Entropy values (as measured by high-througput technologies). At the same time, tumor cells increment their divergence from the normal tissue profile increasing their disorder via creation of states that we might not directly measure. This unifying hallmark allows, via the the Jensen-Shannon divergence, to identify the arrow of time of the processes from the gene expression profiles, and helps to map the phenotypical and molecular hallmarks of specific cancer subtypes. The deep mathematical basis of the approach allows us to suggest that this principle is, hopefully, of general applicability for other diseases.
Collapse
Affiliation(s)
- Regina Berretta
- Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, The University of Newcastle, Callaghan, New South Wales, Australia
- Information Based Medicine Program, Hunter Medical Research Institute, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
| | - Pablo Moscato
- Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, The University of Newcastle, Callaghan, New South Wales, Australia
- Information Based Medicine Program, Hunter Medical Research Institute, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
- Australian Research Council Centre of Excellence in Bioinformatics, Callaghan, New South Wales, Australia
- * E-mail:
| |
Collapse
|
44
|
Abstract
Biomarkers play an important role in the diagnosis and prognostic classification of various cancers and can be useful in monitoring the patient's clinical course of disease and response to therapy. Generally, biomarkers are proteins and their expressions are associated with malignant disease. In the majority of cases, the marker molecules are expressed by the tumour cells themselves or by the tumour microenvironment cells. Thus, most biomarkers can primarily be found in malignant tissues, but after active secretion or passive release at tumour destruction, they become detectable in body fluids such as blood. Besides morphological and histopathological biomarkers (anatomic site, type of the primary tumour, tumour size, invasion depth, vascular invasion and ulceration), an increasing variety of serological markers have been identified, providing the possibility of a more detailed diagnostic and prognostic subgrouping of tumour entities, up to and even changing existing classification systems. The goal of this review is to provide an overview of old and more recent serological biomarkers in malignant melanoma. We will first focus on confirmed and nonconfirmed serum tumour markers, followed by proteomic profiling, an innovative approach to identify new and better serological biomarkers in melanoma, and ending with the predictive factors for treatments in this pathology.
Collapse
|
45
|
Chaiworapongsa T, Romero R, Tarca AL, Kusanovic JP, Gotsch F, Mittal P, Kim SK, Vaisbuch E, Mazaki-Tovi S, Erez O, Dong Z, Kim CJ, Yeo L, Hassan SS. A decrease in maternal plasma concentrations of sVEGFR-2 precedes the clinical diagnosis of preeclampsia. Am J Obstet Gynecol 2010; 202:550.e1-10. [PMID: 20510958 PMCID: PMC3437767 DOI: 10.1016/j.ajog.2010.04.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Revised: 05/27/2009] [Accepted: 04/05/2010] [Indexed: 11/21/2022]
Abstract
OBJECTIVE The aim of this study was to examine if maternal plasma concentrations of soluble vascular endothelial growth factor receptor (sVEGFR)-2 change prior to the diagnosis of preeclampsia. STUDY DESIGN A longitudinal study was conducted in normal pregnant women (n = 160) and patients with preeclampsia (n = 40). Blood samples were collected at 7 gestational age intervals from 6 weeks to term. Plasma concentrations of sVEGFR-2 were determined by enzyme-linked immunosorbent assay. Analysis was performed with cross-sectional and longitudinal (mixed effects model) approaches. RESULTS Mothers destined to develop preeclampsia have lower plasma sVEGFR-2 concentrations than those who will have a normal pregnancy (longitudinal approach; P < .05). Cross-sectional analysis suggested that the median plasma sVEGFR-2 concentration in women destined to develop preeclampsia was significantly lower than that in normal pregnant women from 28-31 weeks of gestation (P = .001) or 6-10 weeks prior to the diagnosis (P < .001). CONCLUSION A lower maternal plasma sVEGFR-2 concentration precedes the development of preeclampsia, both term and preterm.
Collapse
Affiliation(s)
- Tinnakorn Chaiworapongsa
- Perinatology Research Branch, National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Detroit, MI 48201, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Otten J, Bokemeyer C, Fiedler W. Tgf-Beta superfamily receptors-targets for antiangiogenic therapy? JOURNAL OF ONCOLOGY 2010; 2010:317068. [PMID: 20490264 PMCID: PMC2871186 DOI: 10.1155/2010/317068] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Accepted: 02/23/2010] [Indexed: 01/17/2023]
Abstract
The TGF-beta pathway controls a broad range of cellular behavior including cell proliferation, differentiation, and apoptosis of various cell types including tumor cells, endothelial cells, immune cells, and fibroblasts. Besides TGF-beta's direct effects on tumor growth and its involvement in neoangiogenesis have received recent attention. Germline mutations in TGF-beta receptors or coreceptors causing Hereditary Hemorrhagic Teleangiectasia and the Loeys-Dietz syndrome underline the involvement of TGF-beta in vessel formation and maturation. Several therapeutic approaches are evaluated at present targeting the TGF-beta pathway including utilization of antisense oligonucleotides against TGF-beta itself or antibodies or small molecule inhibitors of TGF-beta receptors. Some of these therapeutic agents have already entered the clinical arena including an antibody against the endothelium specific TGF-beta class I receptor ALK-1 targeting tumor vasculature. In conclusion, therapeutic manipulation of the TGF-beta pathway opens great opportunities in future cancer therapy.
Collapse
Affiliation(s)
- Jasmin Otten
- Sections of Pneumonology and Bone Marrow Transplantation, Department of Oncology and Hematology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Carsten Bokemeyer
- Sections of Pneumonology and Bone Marrow Transplantation, Department of Oncology and Hematology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Walter Fiedler
- Sections of Pneumonology and Bone Marrow Transplantation, Department of Oncology and Hematology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
47
|
Aoyagi Y, Iinuma H, Horiuchi A, Shimada R, Watanabe T. Association of plasma VEGF-A, soluble VEGFR-1 and VEGFR-2 levels and clinical response and survival in advanced colorectal cancer patients receiving bevacizumab with modified FOLFOX6. Oncol Lett 2010; 1:253-259. [PMID: 22966290 DOI: 10.3892/ol_00000045] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Accepted: 12/11/2009] [Indexed: 12/19/2022] Open
Abstract
For individualized bevacizumab-based therapy, non-invasive biomarkers are necessary. This study assessed the predictive value of plasma vascular endothelial growth factor (VEGF)-A, soluble VEGF receptor (sVEGFR)-1 and sVEGFR-2 levels as biomarkers for clinical response and survival in advanced colorectal cancer (CRC) patients treated with bevacizumab and modified FOLFOX6 (mFOLFOX6). Forty-six unresectable advanced CRC patients and 20 healthy controls were included in this study. CRC patients were treated with bevacizumab and mFOLFOX6. Pretreatment plasma VEGF-A, sVEGFR-1 and sVEGFR-2 levels were measured using the multiplex immunoassay. Plasma VEGF-A, sVEGFR-1 and sVEGFR-2 levels were significantly higher in CRC patients than in the healthy subjects. The plasma sVEGFR-1 levels in the responder patients [complete response (CR)/partial response (PR)] and stable disease (SD) patients were significantly lower than those in the progressive disease (PD) patients (CR/PR vs. PD, p=0.025; SD vs. PD, p=0.032), while the plasma VEGF-A and sVEGFR-2 levels did not show any significant differences between the two groups of patients. Patients with higher sVEGFR-1 levels showed a significantly poorer progression-free survival (PFS) and overall survival (OS) than those with lower VEGFR-1 levels. In contrast, VEGF-A and sVEGFR-2 did not show any significant relationship between PFS and OS according to the status of each level. In the multivariate Cox proportional hazard regression analysis, sVEGFR-1 levels showed a significant relationship between PFS and OS. These results suggest that plasma sVEGFR-1 levels have a predictive value for clinical response and survival in advanced CRC patients treated with bevacizumab and mFOLFOX6. Larger scale studies are needed to further validate our results.
Collapse
Affiliation(s)
- Yoshiko Aoyagi
- Department of Surgery, Teikyo University School of Medicine, Tokyo 173-0003, Japan
| | | | | | | | | |
Collapse
|
48
|
Mehnert JM, McCarthy MM, Jilaveanu L, Flaherty KT, Aziz S, Camp RL, Rimm DL, Kluger HM. Quantitative expression of VEGF, VEGF-R1, VEGF-R2, and VEGF-R3 in melanoma tissue microarrays. Hum Pathol 2009; 41:375-84. [PMID: 20004943 DOI: 10.1016/j.humpath.2009.08.016] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2009] [Revised: 08/10/2009] [Accepted: 08/18/2009] [Indexed: 12/19/2022]
Abstract
Angiogenesis is required for progression and metastasis of melanoma. Analysis of angiogenic molecules in benign and malignant tissues may allow identification of markers useful for prediction of sensitivity to antiangiogenic agents. We hypothesized that differential expression of vascular endothelial growth factor (VEGF) and its receptors VEGF-R1, VEGF-R2, and VEGF-R3 would be higher in melanomas than nevi and higher in advanced melanoma. Using automated quantitative analysis, we quantified VEGF, -R1, -R2 and -R3 expression in melanoma tissue microarrays composed of 540 nevi and 468 melanoma specimens (198 primaries, 270 metastases). VEGF, VEGF-R1, VEGF-R2, and VEGF-R3 expression was significantly higher in melanomas than nevi by unpaired t tests (P < .0001). VEGF-R2 expression was higher in metastatic specimens (P < .0001), but VEGF-R3 expression was higher in primaries (P < .0001). VEGF was coexpressed with all 3 receptors when assessed by Spearman's rank correlation. VEGF, VEGF-R1, VEGF-R2, and VEGF-R3 expression is higher in melanomas than nevi. Higher expression of VEGF-R2 was found in metastases versus primaries, supporting the idea that selection for an angiogenic phenotype in metastatic melanoma is conferred via up-regulation of VEGF-R2. However, higher expression of VEGF-R3 was seen on primary lesions, potentially implicating this receptor in initiation of lymphatic tumor spread. Clinical trials using antiangiogenic agents in melanoma should include correlative assays of VEGF, VEGF-R1, VEGF-R2, and VEGF-R3 as biomarkers of response to therapy, preferably using quantitative methods such as automated quantitative analysis. Such assessments could assist with evaluation of these molecules as therapeutic targets in melanoma, ultimately facilitating improved selection of patients for treatment.
Collapse
Affiliation(s)
- Janice M Mehnert
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Tripathi R, Rath G, Ralhan R, Saxena S, Salhan S. Soluble and membranous vascular endothelial growth factor receptor-2 in pregnancies complicated by pre-eclampsia. Yonsei Med J 2009; 50:656-66. [PMID: 19881969 PMCID: PMC2768240 DOI: 10.3349/ymj.2009.50.5.656] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 09/02/2008] [Accepted: 12/08/2008] [Indexed: 12/27/2022] Open
Abstract
PURPOSE There is a paucity of information on the serum soluble vascular endothelial growth factor receptor-2 (sVEGFR-2) concentrations, membranous VEGFR-2 expression and the mechanism involved in their modulations during the clinical onset of pre-eclampsia. This cross-sectional study was conducted to evaluate the concentration of sVEGFR-2 in serum and to investigate the expression of membranous VEGFR-2 in placentae of pre-eclampsia group. MATERIALS AND METHODS The serum levels of sVEGFR-2 (n = 120) and the expression of membranous VEGFR-2 in placentae (n = 100) were analysed at third trimester of pregnancy by enzyme linked immunosorbent assay (ELISA) and immunohistochemistry respectively. The diagnostic parameters of sensitivity, specificity and association of soluble and membranous VEGFR-2 in these patients were evaluated. RESULTS The serum levels of sVEGFR-2 in pre-eclampsia patients were found to be significantly reduced (p = 0.01, p = 0.001) in early and late pre-eclamptic sub-groups as compared to their respective third trimester control sub-groups. Also, the receiver operating characteristic (ROC) curve analysis showed a cut-off value of 7350.4 pg/mL, higher sensitivity (76%) and specificity (76%) for sVEGFR-2 in late onset (> 34 weeks) pre-eclamptic group. Significant down-regulation of membranous VEGFR-2 immunoreactivity was observed in all the placental cells (p = 0.0001) at > 34 weeks preeclamptic group. CONCLUSION The reduced serum levels of soluble VEGFR-2 and the down-regulated expression of membranous VEGFR-2 in the study group denoted abnormality in VEGF mediated placental function in all placental cells and thus VEGFR-2 may be a key factor, intimately associated with pre-eclampsia. This study shows the clinical utility of soluble and membranous VEGFR-2 in pre-eclampsia patients.
Collapse
Affiliation(s)
- Richa Tripathi
- Department of Anatomy, VMMC & Safdarjang Hospital, New Delhi, India.
| | | | | | | | | |
Collapse
|
50
|
Mansfield AS, Markovic SN. Novel therapeutics for the treatment of metastatic melanoma. Future Oncol 2009; 5:543-57. [PMID: 19450181 DOI: 10.2217/fon.09.15] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Metastatic malignant melanoma is an incurable disease with a median survival of 8.5 months and a probability of surviving 5 years after the diagnosis of less than 5%. To date, no systemic therapy has meaningfully changed these survival end points. Currently, in the USA the FDA has approved three agents for the treatment of metastatic melanoma: hydroxyurea, dacarbazine and interleukin-2. None of these have demonstrated a meaningfully prolonged survival of patients with metastatic melanoma. Therefore, a number of innovative therapeutic strategies have been pursued to improve outcomes, including immune therapy, tyrosine kinase inhibitors and angiogenesis inhibitors. Herein, we review some of the recent advances in novel therapeutic developments for the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Aaron S Mansfield
- Department of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | | |
Collapse
|