101
|
Functional malignant cell heterogeneity in pancreatic neuroendocrine tumors revealed by targeting of PDGF-DD. Proc Natl Acad Sci U S A 2016; 113:E864-73. [PMID: 26831065 DOI: 10.1073/pnas.1509384113] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Intratumoral heterogeneity is an inherent feature of most human cancers and has profound implications for cancer therapy. As a result, there is an emergent need to explore previously unmapped mechanisms regulating distinct subpopulations of tumor cells and to understand their contribution to tumor progression and treatment response. Aberrant platelet-derived growth factor receptor beta (PDGFRβ) signaling in cancer has motivated the development of several antagonists currently in clinical use, including imatinib, sunitinib, and sorafenib. The discovery of a novel ligand for PDGFRβ, platelet-derived growth factor (PDGF)-DD, opened the possibility of a previously unidentified signaling pathway involved in tumor development. However, the precise function of PDGF-DD in tumor growth and invasion remains elusive. Here, making use of a newly generated Pdgfd knockout mouse, we reveal a functionally important malignant cell heterogeneity modulated by PDGF-DD signaling in pancreatic neuroendocrine tumors (PanNET). Our analyses demonstrate that tumor growth was delayed in the absence of signaling by PDGF-DD. Surprisingly, ablation of PDGF-DD did not affect the vasculature or stroma of PanNET; instead, we found that PDGF-DD stimulated bulk tumor cell proliferation by induction of paracrine mitogenic signaling between heterogeneous malignant cell clones, some of which expressed PDGFRβ. The presence of a subclonal population of tumor cells characterized by PDGFRβ expression was further validated in a cohort of human PanNET. In conclusion, we demonstrate a previously unrecognized heterogeneity in PanNET characterized by signaling through the PDGF-DD/PDGFRβ axis.
Collapse
|
102
|
Ferrara N, Adamis AP, Albeck M, Sredni B. Ten years of anti-vascular endothelial growth factor therapy. Nat Rev Drug Discov 2016; 15:385-403. [PMID: 26775688 DOI: 10.1038/nrd.2015.17] [Citation(s) in RCA: 662] [Impact Index Per Article: 82.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The targeting of vascular endothelial growth factor A (VEGFA), a crucial regulator of both normal and pathological angiogenesis, has revealed innovative therapeutic approaches in oncology and ophthalmology. The first VEGFA inhibitor, bevacizumab, was approved by the US Food and Drug Administration in 2004 for the first-line treatment of metastatic colorectal cancer, and the first VEGFA inhibitors in ophthalmology, pegaptanib and ranibizumab, were approved in 2004 and 2006, respectively. To mark this tenth anniversary of anti-VEGFA therapy, we discuss the discovery of VEGFA, the successes and challenges in the development of VEGFA inhibitors and the impact of these agents on the treatment of cancers and ophthalmic diseases.
Collapse
Affiliation(s)
- Napoleone Ferrara
- University of California, San Diego, La Jolla, California, 92093, USA
| | | | | | | |
Collapse
|
103
|
Drolet DW, Green LS, Gold L, Janjic N. Fit for the Eye: Aptamers in Ocular Disorders. Nucleic Acid Ther 2016; 26:127-46. [PMID: 26757406 PMCID: PMC4900223 DOI: 10.1089/nat.2015.0573] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
For any new class of therapeutics, there are certain types of indications that represent a natural fit. For nucleic acid ligands in general, and aptamers in particular, the eye has historically been an attractive site for therapeutic intervention. In this review, we recount the discovery and early development of three aptamers designated for use in ophthalmology, one approved (Macugen), and two in late-stage development (Fovista and Zimura). Every one of these molecules was originally intended for other indications. Key improvements in technology, specifically with regard to libraries used for in vitro selection and subsequent chemical optimization of aptamers, have played an important role in allowing the identification of development candidates with suitable properties. The lessons learned from the selection of these molecules are valuable for informing us about the many remaining opportunities for aptamer-based therapeutics in ophthalmology as well as for identifying additional indications for which aptamers as a class of therapeutics have distinct advantages.
Collapse
|
104
|
Vascular Wall-Resident Multipotent Stem Cells of Mesenchymal Nature within the Process of Vascular Remodeling: Cellular Basis, Clinical Relevance, and Implications for Stem Cell Therapy. Stem Cells Int 2016; 2016:1905846. [PMID: 26880936 PMCID: PMC4736960 DOI: 10.1155/2016/1905846] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/18/2015] [Indexed: 12/13/2022] Open
Abstract
Until some years ago, the bone marrow and the endothelial cell compartment lining the vessel lumen (subendothelial space) were thought to be the only sources providing vascular progenitor cells. Now, the vessel wall, in particular, the vascular adventitia, has been established as a niche for different types of stem and progenitor cells with the capacity to differentiate into both vascular and nonvascular cells. Herein, vascular wall-resident multipotent stem cells of mesenchymal nature (VW-MPSCs) have gained importance because of their large range of differentiation in combination with their distribution throughout the postnatal organism which is related to their existence in the adventitial niche, respectively. In general, mesenchymal stem cells, also designated as mesenchymal stromal cells (MSCs), contribute to the maintenance of organ integrity by their ability to replace defunct cells or secrete cytokines locally and thus support repair and healing processes of the affected tissues. This review will focus on the central role of VW-MPSCs within vascular reconstructing processes (vascular remodeling) which are absolute prerequisite to preserve the sensitive relationship between resilience and stability of the vessel wall. Further, a particular advantage for the therapeutic application of VW-MPSCs for improving vascular function or preventing vascular damage will be discussed.
Collapse
|
105
|
Abstract
Resistance to chemotherapy is among the most important issues in the management of ovarian cancer. Unlike cancer cells, which are heterogeneous as a result of remarkable genetic instability, stromal cells are considered relatively homogeneous. Thus, targeting the tumor microenvironment is an attractive approach for cancer therapy. Arguably, anti-vascular endothelial growth factor (anti-VEGF) therapies hold great promise, but their efficacy has been modest, likely owing to redundant and complementary angiogenic pathways. Components of platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), epidermal growth factor (EGF), and other pathways may compensate for VEGF blockade and allow angiogenesis to occur despite anti-VEGF treatment. In addition, hypoxia induced by anti-angiogenesis therapy modifies signaling pathways in tumor and stromal cells, which induces resistance to therapy. Because of tumor cell heterogeneity and angiogenic pathway redundancy, combining cytotoxic and targeted therapies or combining therapies targeting different pathways can potentially overcome resistance. Although targeted therapy is showing promise, much more work is needed to maximize its impact, including the discovery of new targets and identification of individuals most likely to benefit from such therapies.
Collapse
|
106
|
Bhome R, Al Saihati H, Goh R, Bullock M, Primrose J, Thomas G, Sayan A, Mirnezami A. Translational aspects in targeting the stromal tumour microenvironment: from bench to bedside. NEW HORIZONS IN TRANSLATIONAL MEDICINE 2016; 3:9-21. [PMID: 27275004 PMCID: PMC4888939 DOI: 10.1016/j.nhtm.2016.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 03/07/2016] [Accepted: 03/24/2016] [Indexed: 02/07/2023]
Abstract
Solid tumours comprise, not only malignant cells but also a variety of stromal cells and extracellular matrix proteins. These components interact via an array of signalling pathways to create an adaptable network that may act to promote or suppress cancer progression. To date, the majority of anti-tumour chemotherapeutic agents have principally sought to target the cancer cell. Consequently, resistance develops because of clonal evolution, as a result of selection pressure during tumour expansion. The concept of activating or inhibiting other cell types within the tumour microenvironment is relatively novel and has the advantage of targeting cells which are genetically stable and less likely to develop resistance. This review outlines key players in the stromal tumour microenvironment and discusses potential targeting strategies that may offer therapeutic benefit.
Collapse
Affiliation(s)
- R. Bhome
- Cancer Sciences, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
- University Surgery, South Academic Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - H.A. Al Saihati
- Cancer Sciences, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - R.W. Goh
- Cancer Sciences, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
- School of Medicine, University of Southampton, University Road, Southampton SO17 1BJ, UK
| | - M.D. Bullock
- Cancer Sciences, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
- University Surgery, South Academic Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - J.N. Primrose
- University Surgery, South Academic Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - G.J. Thomas
- Cancer Sciences, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - A.E. Sayan
- Cancer Sciences, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - A.H. Mirnezami
- Cancer Sciences, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
- University Surgery, South Academic Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| |
Collapse
|
107
|
Mitamura T, Gourley C, Sood AK. Prediction of anti-angiogenesis escape. Gynecol Oncol 2015; 141:80-5. [PMID: 26748214 DOI: 10.1016/j.ygyno.2015.12.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/17/2015] [Accepted: 12/30/2015] [Indexed: 01/29/2023]
Abstract
Many clinical trials have demonstrated the benefit of anti-angiogenesis therapy in the treatment of gynecologic cancer. However, these benefits have often been in terms of progression-free rather than overall survival and in some cases, the magnitude of benefit demonstrated in the pivotal phase 3 trials has been disappointing when compared with the percentage of patients who responded in earlier phase 2 trials. Two potential explanations for this are the current inability to stratify patients according to chance of benefit and the development of resistance mechanisms within the tumor. In this article, we review the prediction of response and the proposed resistance and escape mechanisms involved in anti-angiogenesis therapy, including the up-regulation of alternative proangiogenic pathways, vascular co-option, and resistance to hypoxia. These insights may offer a personalized strategy for anti-angiogenesis therapy and help us to consider the best selection of other therapies that should be combined with anti-angiogenesis therapy to improve the outcome of patients with gynecologic cancer.
Collapse
Affiliation(s)
- Takashi Mitamura
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030, USA.
| | - Charlie Gourley
- University of Edinburgh Cancer Research UK Centre, MRC IGMM, Crewe Road South, Edinburgh, EH4 2XR, UK.
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
108
|
Awasthi N, Schwarz RE. Profile of nintedanib in the treatment of solid tumors: the evidence to date. Onco Targets Ther 2015; 8:3691-701. [PMID: 26677336 PMCID: PMC4677757 DOI: 10.2147/ott.s78805] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Angiogenesis is an essential process for tumor growth and metastasis, and remains a promising therapeutic target process in cancer treatment for several cancer types. Bevacizumab, a monoclonal antibody that targets vascular endothelial growth factor (VEGF), was the first antiangiogenic agent approved for cancer therapy. Novel antiangiogenic agents, such as sunitinib, sorafenib, pazopanib, or vandetanib that target additional proangiogenic signaling pathways beyond VEGF, have also been approved for the treatment of various malignant diseases. While most of these agents are approved in combination with cytotoxic chemotherapy for indications including metastatic colorectal cancer, non-small-cell lung cancer, breast cancer, renal cell carcinoma (RCC), and gastric cancer, some are used as approved monotherapy for advanced RCC, hepatocellular carcinoma and medullary thyroid cancer. Major challenges to the success of antiangiogenic therapy include associated toxicity risks, limitation of efficacy through the possible development of resistance and induction or promotion of metastatic progression. Nintedanib (formally known as BIBF 1120) is a triple angiokinase inhibitor of VEGF, fibroblast growth factor, platelet-derived growth factor signaling with lesser activity against RET, Flt-3, and Src. Through this unique targeting profile nintedanib has demonstrated significant antitumor activity in several tumor types in preclinical studies. Nintedanib has also shown promising clinical efficacy in combination with docetaxel and has been approved for treating patients with locally advanced and metastatic non-small-cell lung cancer in Europe. Nintedanib has also been found to be clinically promising in terms of efficacy and safety in several other solid tumors including ovarian cancer (Phase III), RCC (Phase II), and prostate cancer (Phase II). This review article provides a comprehensive summary of the preclinical and clinical efficacy of nintedanib in the treatment of solid tumors.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, USA
| | - Roderich E Schwarz
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, USA ; Indiana University Health Goshen Center for Cancer Care, Goshen, IN, USA
| |
Collapse
|
109
|
Fibrocyte-like cells mediate acquired resistance to anti-angiogenic therapy with bevacizumab. Nat Commun 2015; 6:8792. [PMID: 26635184 PMCID: PMC4686833 DOI: 10.1038/ncomms9792] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 10/02/2015] [Indexed: 12/19/2022] Open
Abstract
Bevacizumab exerts anti-angiogenic effects in cancer patients by inhibiting vascular endothelial growth factor (VEGF). However, its use is still limited due to the development of resistance to the treatment. Such resistance can be regulated by various factors, although the underlying mechanisms remain incompletely understood. Here we show that bone marrow-derived fibrocyte-like cells, defined as alpha-1 type I collagen-positive and CXCR4-positive cells, contribute to the acquired resistance to bevacizumab. In mouse models of malignant pleural mesothelioma and lung cancer, fibrocyte-like cells mediate the resistance to bevacizumab as the main producer of fibroblast growth factor 2. In clinical specimens of lung cancer, the number of fibrocyte-like cells is significantly increased in bevacizumab-treated tumours, and correlates with the number of treatment cycles, as well as CD31-positive vessels. Our results identify fibrocyte-like cells as a promising cell biomarker and a potential therapeutic target to overcome resistance to anti-VEGF therapy.
Collapse
|
110
|
Mayo JN, Bearden SE. Driving the Hypoxia-Inducible Pathway in Human Pericytes Promotes Vascular Density in an Exosome-Dependent Manner. Microcirculation 2015; 22:711-23. [PMID: 26243428 PMCID: PMC4715585 DOI: 10.1111/micc.12227] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 07/29/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The mechanisms involved in activating pericytes, cells that ensheath capillaries, to engage in the formation of new capillaries, angiogenesis, remain unknown. In this study, the hypothesis was tested that pericytes could be stimulated to promote angiogenesis by driving the HIF pathway. METHODS Pericytes were stimulated with CoCl2 to activate the HIF pathway. Stimulated pericytes were cocultured with endothelial cells in a wound healing assay and in a 3D collagen matrix assay of angiogenesis. A culture system of spinal cord tissue was used to assess microvascular outcomes after treatment with stimulated pericytes. Pharmaceutical inhibition of exosome production was also performed. RESULTS Treatment with stimulated pericytes resulted in faster wound healing (1.92 ± 0.18 fold increase, p < 0.05), greater endothelial cord formation (2.9 ± 0.14 fold increase, p < 0.05) in cell culture assays, and greater vascular density (1.78 ± 0.23 fold increase, p < 0.05) in spinal cord tissue. Exosome secretion and the physical presence of stimulated pericytes were necessary in the promotion of angiogenic outcomes. CONCLUSIONS These results elucidate a mechanism that may be exploited to enhance features of angiogenesis in the CNS.
Collapse
Affiliation(s)
- Jamie N. Mayo
- Department of Biological Sciences, Idaho State University, 921 S. 8th Ave Stop 8007, Pocatello, ID, 83209
| | - Shawn E. Bearden
- Department of Biological Sciences, Idaho State University, 921 S. 8th Ave Stop 8007, Pocatello, ID, 83209
- ISU Biomedical Research Institute, Idaho State University, 921 S. 8th Ave Stop 8046, Pocatello, ID, 83209
| |
Collapse
|
111
|
Huang YM, Wang CH, Huang JS, Yeh KY, Lai CH, Wu TH, Chang PH, Chang PH, Chang YS, Lan YJ. Dasatinib-related chylothorax. Turk J Haematol 2015; 32:68-72. [PMID: 25805678 PMCID: PMC4439910 DOI: 10.4274/tjh.2012.0196] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Dasatinib is a potent second-generation tyrosine kinase inhibitor for the treatment of chronic myeloid leukemia. The most common adverse event associated with dasatinib therapy is fluid retention, including pleural effusion. Dasatinib-related chylothorax has rarely been reported. The clinical manifestations, pathophysiology, management, and prognosis are not fully understood. Here we report a 40-year-old woman presenting with chylothorax following dasatinib use. We propose the hypothesis of its mechanism as well as offering a review of the relevant literature.
Collapse
Affiliation(s)
- Yen Min Huang
- Chang Gung Memorial Hospital, Clinic of Internal Medicine, Division of Hematology-Oncology, Keelung, Taiwan. E-mail:
| | | | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Sadanandam A, Wullschleger S, Lyssiotis CA, Grötzinger C, Barbi S, Bersani S, Körner J, Wafy I, Mafficini A, Lawlor RT, Simbolo M, Asara JM, Bläker H, Cantley LC, Wiedenmann B, Scarpa A, Hanahan D. A Cross-Species Analysis in Pancreatic Neuroendocrine Tumors Reveals Molecular Subtypes with Distinctive Clinical, Metastatic, Developmental, and Metabolic Characteristics. Cancer Discov 2015; 5:1296-313. [PMID: 26446169 DOI: 10.1158/2159-8290.cd-15-0068] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 10/05/2015] [Indexed: 12/14/2022]
Abstract
UNLABELLED Seeking to assess the representative and instructive value of an engineered mouse model of pancreatic neuroendocrine tumors (PanNET) for its cognate human cancer, we profiled and compared mRNA and miRNA transcriptomes of tumors from both. Mouse PanNET tumors could be classified into two distinctive subtypes, well-differentiated islet/insulinoma tumors (IT) and poorly differentiated tumors associated with liver metastases, dubbed metastasis-like primary (MLP). Human PanNETs were independently classified into these same two subtypes, along with a third, specific gene mutation-enriched subtype. The MLP subtypes in human and mouse were similar to liver metastases in terms of miRNA and mRNA transcriptome profiles and signature genes. The human/mouse MLP subtypes also similarly expressed genes known to regulate early pancreas development, whereas the IT subtypes expressed genes characteristic of mature islet cells, suggesting different tumorigenesis pathways. In addition, these subtypes exhibit distinct metabolic profiles marked by differential pyruvate metabolism, substantiating the significance of their separate identities. SIGNIFICANCE This study involves a comprehensive cross-species integrated analysis of multi-omics profiles and histology to stratify PanNETs into subtypes with distinctive characteristics. We provide support for the RIP1-TAG2 mouse model as representative of its cognate human cancer with prospects to better understand PanNET heterogeneity and consider future applications of personalized cancer therapy.
Collapse
Affiliation(s)
- Anguraj Sadanandam
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland. Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Lausanne (EPFL), Lausanne, Switzerland. Division of Molecular Pathology, Institute of Cancer Research (ICR), London, United Kingdom.
| | - Stephan Wullschleger
- Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Lausanne (EPFL), Lausanne, Switzerland
| | | | - Carsten Grötzinger
- Department of Hepatology and Gastroenterology, Charite, Campus Virchow-Klinikum, University Medicine Berlin, Berlin, Germany
| | - Stefano Barbi
- ARC-Net Research Centre and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Samantha Bersani
- ARC-Net Research Centre and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Jan Körner
- Department of Hepatology and Gastroenterology, Charite, Campus Virchow-Klinikum, University Medicine Berlin, Berlin, Germany
| | - Ismael Wafy
- Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Lausanne (EPFL), Lausanne, Switzerland
| | - Andrea Mafficini
- ARC-Net Research Centre and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Rita T Lawlor
- ARC-Net Research Centre and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Michele Simbolo
- ARC-Net Research Centre and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - John M Asara
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Hendrik Bläker
- Institut für Pathologie, Charite, Campus Virchow-Klinikum, University Medicine, Berlin, Germany
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medical College, New York, New York
| | - Bertram Wiedenmann
- Department of Hepatology and Gastroenterology, Charite, Campus Virchow-Klinikum, University Medicine Berlin, Berlin, Germany
| | - Aldo Scarpa
- ARC-Net Research Centre and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy.
| | - Douglas Hanahan
- Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
113
|
Rodriguez-Freixinos V, Mackay HJ. Breaking down the evidence for bevacizumab in advanced cervical cancer: past, present and future. GYNECOLOGIC ONCOLOGY RESEARCH AND PRACTICE 2015; 2:8. [PMID: 27231568 PMCID: PMC4881045 DOI: 10.1186/s40661-015-0015-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/16/2015] [Indexed: 12/15/2022]
Abstract
Despite the introduction of screening and, latterly, vaccination programs in the developed world, globally cervical cancer remains a significant health problem. For those diagnosed with advanced or recurrent disease even within resource rich communities, prognosis remains poor with an overall survival (OS) of just over 12 months. New therapeutic interventions are urgently required. Advances in our understanding of the mechanisms underlying tumor growth and the downstream effects of human papilloma virus (HPV) infection identified angiogenesis as a rational target for therapeutic intervention in cervical cancer. Anti-angiogenic agents showed promising activity in early phase clinical trials culminating in a randomized phase III study of the humanized monoclonal antibody to vascular endothelial growth factor (VEGF), bevacizumab, in combination with chemotherapy. This pivotal study, the Gynecologic Oncology Group protocol 240, met its primary endpoint demonstrating a significant improvement in OS. Bevacizumab became the first targeted agent to be granted regulatory approval by the United States Food and Drug Administration for use alongside chemotherapy in adults with persistent, recurrent or metastatic carcinoma of the cervix. This review outlines the rationale for targeting angiogenesis in cervical cancer focusing on the current indications for the use of bevacizumab in this disease and future directions.
Collapse
Affiliation(s)
- Victor Rodriguez-Freixinos
- Division of Medical Oncology and Hematology, Princess Margaret Hospital, University of Toronto, 610 University Avenue, Toronto, Ontario M5G 2 M9 Canada
| | - Helen J. Mackay
- Division of Medical Oncology and Hematology, Princess Margaret Hospital, University of Toronto, 610 University Avenue, Toronto, Ontario M5G 2 M9 Canada
| |
Collapse
|
114
|
The vascular disrupting activity of OXi8006 in endothelial cells and its phosphate prodrug OXi8007 in breast tumor xenografts. Cancer Lett 2015; 369:229-41. [PMID: 26325604 DOI: 10.1016/j.canlet.2015.08.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 08/24/2015] [Accepted: 08/24/2015] [Indexed: 12/31/2022]
Abstract
This study describes the vascular disrupting ability and the mechanism of action of the indole-based tubulin-binding compound, OXi8006, and its water-soluble phosphate prodrug OXi8007. Treatment of rapidly proliferating human umbilical vein endothelial cells (HUVECs), used as a model for the tumor vasculature, with OXi8006 or OXi8007, caused potent microtubule disruption followed by extensive reorganization of the cytoskeletal network. The mechanism of action involved an increase in focal adhesion formation associated with an increase in phosphorylation of both non-muscle myosin light chain and focal adhesion kinase. These effects were dramatically diminished by an inhibitor of RhoA kinase, a downstream effector of RhoA. Cell cycle blockade at G2/M and cytotoxicity toward rapidly proliferating HUVECs were also observed. Capillary-like networks of HUVECs were disrupted by the action of both OXi8006 and OXi8007. The prodrug OXi8007 exhibited potent and rapid dose-dependent antivascular activity assessed by dynamic bioluminescence imaging (BLI) in an MDA-MB-231-luc breast cancer xenograft mouse model. By 6 hours post treatment, over 93% of the BLI signal was abolished with only a slight recovery at 24 hours. These findings were confirmed by histology. The results from this study demonstrate that OXi8007 is a potent vascular disrupting agent acting through an anti-microtubule mechanism involving RhoA.
Collapse
|
115
|
Langlois B, Saupe F, Rupp T, Arnold C, van der Heyden M, Orend G, Hussenet T. AngioMatrix, a signature of the tumor angiogenic switch-specific matrisome, correlates with poor prognosis for glioma and colorectal cancer patients. Oncotarget 2015; 5:10529-45. [PMID: 25301723 PMCID: PMC4279391 DOI: 10.18632/oncotarget.2470] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 09/06/2014] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis represents a rate-limiting step during tumor progression. Targeting angiogenesis is already applied in cancer treatment, yet limits of anti-angiogenic therapies have emerged, notably because tumors adapt and recur after treatment. Therefore, there is a strong need to better understand the molecular and cellular mechanisms underlying tumor angiogenesis. Using the RIP1-Tag2 transgenic murine model, we identified 298 genes that are deregulated during the angiogenic switch, revealing an ingression/expansion of specific stromal cell types including endothelial cells and pericytes, but also macrophages and perivascular mesenchymal cells. Canonical TGF-β signaling is up-regulated during the angiogenic switch, especially in tumor-associated macrophages and fibroblasts. The matrisome, comprising extracellular matrix (ECM) and ECM-associated molecules, is significantly enriched, which allowed us to define the AngioMatrix signature as the 110 matrisomal genes induced during the RIP1-Tag2 angiogenic switch. Several AngioMatrix molecules were validated at expression level. Ablation of tenascin-C, one of the most highly induced ECM molecules during the switch, resulted in reduced angiogenesis confirming its important role. In human glioma and colorectal samples, the AngioMatrix signature correlates with the expression of endothelial cell markers, is increased with tumor progression and finally correlates with poor prognosis demonstrating its diagnostic and therapeutic potential.
Collapse
Affiliation(s)
- Benoit Langlois
- Inserm U1109, MN3T team, Molière, Strasbourg, 67200, France. Université de Strasbourg, Strasbourg, 67000, France. LabEx Medalis, Université de Strasbourg, Strasbourg, 67000, France. Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, 67000, France
| | - Falk Saupe
- Inserm U1109, MN3T team, Molière, Strasbourg, 67200, France. Université de Strasbourg, Strasbourg, 67000, France. LabEx Medalis, Université de Strasbourg, Strasbourg, 67000, France. Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, 67000, France
| | - Tristan Rupp
- Inserm U1109, MN3T team, Molière, Strasbourg, 67200, France. Université de Strasbourg, Strasbourg, 67000, France. LabEx Medalis, Université de Strasbourg, Strasbourg, 67000, France. Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, 67000, France
| | - Christiane Arnold
- Inserm U1109, MN3T team, Molière, Strasbourg, 67200, France. Université de Strasbourg, Strasbourg, 67000, France. LabEx Medalis, Université de Strasbourg, Strasbourg, 67000, France. Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, 67000, France
| | - Michaël van der Heyden
- Inserm U1109, MN3T team, Molière, Strasbourg, 67200, France. Université de Strasbourg, Strasbourg, 67000, France. LabEx Medalis, Université de Strasbourg, Strasbourg, 67000, France. Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, 67000, France
| | - Gertraud Orend
- Inserm U1109, MN3T team, Molière, Strasbourg, 67200, France. Université de Strasbourg, Strasbourg, 67000, France. LabEx Medalis, Université de Strasbourg, Strasbourg, 67000, France. Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, 67000, France
| | - Thomas Hussenet
- Inserm U1109, MN3T team, Molière, Strasbourg, 67200, France. Université de Strasbourg, Strasbourg, 67000, France. LabEx Medalis, Université de Strasbourg, Strasbourg, 67000, France. Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, 67000, France
| |
Collapse
|
116
|
Correa D, Somoza RA, Lin P, Schiemann WP, Caplan AI. Mesenchymal stem cells regulate melanoma cancer cells extravasation to bone and liver at their perivascular niche. Int J Cancer 2015; 138:417-27. [PMID: 26235173 DOI: 10.1002/ijc.29709] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 07/08/2015] [Accepted: 07/20/2015] [Indexed: 12/25/2022]
Abstract
Skeleton and liver are preferred organs for cancer dissemination in metastatic melanoma negatively impacting quality of life, therapeutic success and overall survival rates. At the target organ, the local microenvironment and cell-to-cell interactions between invading and resident stromal cells constitute critical components during the establishment and progression of metastasis. Mesenchymal stem cells (MSCs) possess, in addition to their cell progenitor function, a secretory capacity based on cooperativity with other cell types in injury sites including primary tumors (PT). However, their role at the target organ microenvironment during cancer dissemination is not known. We report that local MSCs, acting as pericytes, regulate the extravasation of melanoma cancer cells (MCC) specifically to murine bone marrow (BM) and liver. Intra-arterially injected wild-type MCC fail to invade those selective organs in a genetic model of perturbed pericyte coverage of the vasculature (PDGF-B(ret/ret)), similar to CD146-deficient MCC injected into wild type mice. Invading MCC interact with resident MSCs/pericytes at the perivascular space through co-expressed CD146 and Sdf-1/CXCL12-CXCR4 signaling. Implanted engineered bone structures with MSCs/pericytes deficient of either Sdf-1/CXCL12 or CD146 become resistant to invasion by circulating MCC. Collectively, the presence of MSCs/pericytes surrounding the target organ vasculature is required for efficient melanoma metastasis to BM and liver.
Collapse
Affiliation(s)
- Diego Correa
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, OH
| | - Rodrigo A Somoza
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, OH
| | - Paul Lin
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, OH
| | - William P Schiemann
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Arnold I Caplan
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
117
|
Flanigan PM, Aghi MK. Adaptation to antiangiogenic therapy in neurological tumors. Cell Mol Life Sci 2015; 72:3069-82. [PMID: 25943307 PMCID: PMC4506875 DOI: 10.1007/s00018-015-1916-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 04/21/2015] [Accepted: 04/23/2015] [Indexed: 01/15/2023]
Abstract
Because tumors require a vascular supply for their survival and growth, angiogenesis is considered an important therapeutic target in most human cancers including cancer of the central nervous system. Antiangiogenic therapy has focused on inhibitors of the vascular endothelial growth factor (VEGF) signaling pathway. VEGF pathway-targeted drugs have shown therapeutic efficacy in several CNS tumors and have been tried most frequently in glioblastoma. These therapies, however, have been less effective than anticipated as some patients do not respond to therapy and some receive only modest benefit. Underlying this suboptimal response are multiple mechanisms of drug resistance involving changes in both tumor cells and their microenvironment. In this review, we discuss the multiple proposed mechanisms by which neurological tumors evolve to become resistant to antiangiogenic therapies. A better understanding of these mechanisms, their context, and their interplay will likely facilitate improvements in pharmacological strategies for the targeted treatment of neurological tumors.
Collapse
Affiliation(s)
| | - Manish K. Aghi
- Department of Neurological Surgery, California Center for Pituitary Disorders, University of California, San Francisco, USA
| |
Collapse
|
118
|
Abstract
Pericytes enveloping the endothelium play an important role in the physiology and pathology of microvessels, especially in vessel maturation and stabilization. However, our understanding of fundamental pericyte biology is limited by the lack of a robust in vitro model system that allows researchers to evaluate the interactions among multiple cell types in perfusable blood vessels. The present work describes a microfluidic platform that can be used to investigate interactions between pericytes and endothelial cells (ECs) during the sprouting, growth, and maturation steps of neovessel formation. A mixture of ECs and pericytes was attached to the side of a pre-patterned three dimensional fibrin matrix and allowed to sprout across the matrix. The effects of intact coverage and EC maturation by the pericytes on the perfused EC network were confirmed using a confocal microscope. Compared with EC monoculture conditions, EC-pericyte co-cultured vessels showed a significant reduction in diameter, increased numbers of junctions and branches and decreased permeability. In response to biochemical factors, ECs and pericytes in the platform showed the similar features with previous reports from in vivo experiments, thus reflect various pathophysiological conditions of in vivo microvessels. Taken together, these results support the physiological relevancy of our three-dimensional microfluidic culture system but also that the system can be used to screen drug effect on EC-pericyte biology.
Collapse
Affiliation(s)
- Jaerim Kim
- Division of WCU Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea
| | - Minhwan Chung
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea
| | - Sudong Kim
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea
| | - Dong Hyun Jo
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Biomedical Sciences and Protein Metabolism Medical Research Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jeong Hun Kim
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Biomedical Sciences and Protein Metabolism Medical Research Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Department of Ophthalmology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- * E-mail: (NLJ); (JHK)
| | - Noo Li Jeon
- Division of WCU Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea
- Institute of Advanced Machinery and Design, Seoul National University, Seoul, Republic of Korea
- * E-mail: (NLJ); (JHK)
| |
Collapse
|
119
|
Flaherty KT, Hamilton BK, Rosen MA, Amaravadi RK, Schuchter LM, Gallagher M, Chen H, Sehgal C, O'Dwyer PJ. Phase I/II Trial of Imatinib and Bevacizumab in Patients With Advanced Melanoma and Other Advanced Cancers. Oncologist 2015; 20:952-9. [PMID: 26084808 DOI: 10.1634/theoncologist.2015-0108] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/21/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Vascular endothelial growth factor and platelet-derived growth factor signaling in the tumor microenvironment appear to cooperate in promoting tumor angiogenesis. PATIENTS AND METHODS We conducted a phase I trial combining bevacizumab (i.v. every 2 weeks) and imatinib (oral daily). Once a recommended phase II dose combination was established, a phase II trial was initiated in patients with metastatic melanoma. A Simon 2-stage design was used with 23 patients required in the first stage and 41 patients in total should the criteria to proceed be met. We required that 50% of the patients be progression-free at 16 weeks. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and power Doppler ultrasonography were performed in patients with metastatic tumors amenable to imaging with these methods at baseline and after 4 weeks. RESULTS A total of 17 patients were accrued to 4 dose and combination levels. Bevacizumab 10 mg/kg every 2 weeks could be safely combined with imatinib 800 mg daily. Common toxicities included fatigue, nausea, vomiting, edema, proteinuria, and anemia, but were not commonly severe. A total of 23 patients with metastatic melanoma (48% with American Joint Commission on Cancer stage M1c; median age, 63 years) were enrolled in the first stage of phase II. The 16-week progression-free survival rate was 35%, leading to termination of phase II after the first stage. In the small subset of patients who remained on study with lesions evaluable by DCE-MRI, significant decreases in tumor vascular permeability were noted, despite early disease progression using the Response Evaluation Criteria In Solid Tumors. CONCLUSION Bevacizumab and imatinib can be safely combined at the maximum doses used for each agent. We did not observe significant clinical activity with this regimen in melanoma patients.
Collapse
Affiliation(s)
- Keith T Flaherty
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Betty K Hamilton
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark A Rosen
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Ravi K Amaravadi
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Lynn M Schuchter
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Maryann Gallagher
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Helen Chen
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Chandra Sehgal
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Peter J O'Dwyer
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
120
|
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, and can be further classified as nonsquamous carcinoma (including adenocarcinoma, which accounts for 40 % of NSCLCs) and squamous NSCLC, which makes up 30 % of NSCLC cases. The emergence of inhibitors of epidermal growth factor receptors, anaplastic lymphoma kinase, and vascular endothelial growth factors (VEGF) in the last decade has resulted in steady improvement in clinical outcomes for patients with advanced lung adenocarcinoma. However, improvements in the survival of patients with squamous NSCLC have remained elusive, presenting an urgent need for understanding and investigating therapeutically relevant molecular targets, specifically in squamous NSCLC. Although anti-VEGF therapy has been studied in squamous NSCLC, progress has been slow, in part due to issues related to pulmonary hemorrhage. In addition to these safety concerns, several phase III trials that initially included patients with squamous NSCLC failed to demonstrate improved overall survival (primary endpoint) with the addition of antiangiogenic therapy to chemotherapy compared with chemotherapy alone. Angiogenesis is an established hallmark of tumor progression and metastasis, and the role of VEGF signaling in angiogenesis is well established. However, some studies suggest that, while inhibiting VEGF signaling may be beneficial, prolonged exposure to VEGF/VEGF receptor (VEGFR) inhibitors may allow tumor cells to utilize alternative angiogenic mechanisms and become resistant. As a result, agents that target multiple angiogenic pathways simultaneously are also under evaluation. This review focuses on current and investigational antiangiogenic targets in squamous NSCLC, including VEGF/VEGFRs, fibroblast growth factor receptors, platelet-derived growth factor receptors, and angiopoietin. Additionally, clinical trials investigating VEGF- and multi-targeted antiangiogenic therapies are discussed.
Collapse
Affiliation(s)
- Bilal Piperdi
- Montefiore Medical Center, 1300 Morris Park Avenue, Mazur Building, Room 616, Bronx, NY, 10461, USA,
| | | | | |
Collapse
|
121
|
Zhao Y, Adjei AA. Targeting Angiogenesis in Cancer Therapy: Moving Beyond Vascular Endothelial Growth Factor. Oncologist 2015; 20:660-73. [PMID: 26001391 DOI: 10.1634/theoncologist.2014-0465] [Citation(s) in RCA: 408] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/06/2015] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Angiogenesis, or the formation of new capillary blood vessels, occurs primarily during human development and reproduction; however, aberrant regulation of angiogenesis is also a fundamental process found in several pathologic conditions, including cancer. As a process required for invasion and metastasis, tumor angiogenesis constitutes an important point of control of cancer progression. Although not yet completely understood, the complex process of tumor angiogenesis involves highly regulated orchestration of multiple signaling pathways. The proangiogenic signaling molecule vascular endothelial growth factor (VEGF) and its cognate receptor (VEGF receptor 2 [VEGFR-2]) play a central role in angiogenesis and often are highly expressed in human cancers, and initial clinical efforts to develop antiangiogenic treatments focused largely on inhibiting VEGF/VEGFR signaling. Such approaches, however, often lead to transient responses and further disease progression because angiogenesis is regulated by multiple pathways that are able to compensate for each other when single pathways are inhibited. The platelet-derived growth factor (PDGF) and PDGF receptor (PDGFR) and fibroblast growth factor (FGF) and FGF receptor (FGFR) pathways, for example, provide potential escape mechanisms from anti-VEGF/VEGFR therapy that could facilitate resumption of tumor growth. Accordingly, more recent treatments have focused on inhibiting multiple signaling pathways simultaneously. This comprehensive review discusses the limitations of inhibiting VEGF signaling alone as an antiangiogenic strategy, the importance of other angiogenic pathways including PDGF/PDGFR and FGF/FGFR, and the novel current and emerging agents that target multiple angiogenic pathways for the treatment of advanced solid tumors. IMPLICATIONS FOR PRACTICE Significant advances in cancer treatment have been achieved with the development of antiangiogenic agents, the majority of which have focused on inhibition of the vascular endothelial growth factor (VEGF) pathway. VEGF targeting alone, however, has not proven to be as efficacious as originally hoped, and it is increasingly clear that there are many interconnected and compensatory pathways that can overcome VEGF-targeted inhibition of angiogenesis. Maximizing the potential of antiangiogenic therapy is likely to require a broader therapeutic approach using a new generation of multitargeted antiangiogenic agents.
Collapse
Affiliation(s)
- Yujie Zhao
- Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Alex A Adjei
- Roswell Park Cancer Institute, Buffalo, New York, USA
| |
Collapse
|
122
|
Combined effects of pericytes in the tumor microenvironment. Stem Cells Int 2015; 2015:868475. [PMID: 26000022 PMCID: PMC4427118 DOI: 10.1155/2015/868475] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/28/2015] [Indexed: 12/25/2022] Open
Abstract
Pericytes are multipotent perivascular cells whose involvement in vasculature development is well established. Evidences in the literature also suggest that pericytes display immune properties and that these cells may serve as an in vivo reservoir of stem cells, contributing to the regeneration of diverse tissues. Pericytes are also capable of tumor homing and are important cellular components of the tumor microenvironment (TME). In this review, we highlight the contribution of pericytes to some classical hallmarks of cancer, namely, tumor angiogenesis, growth, metastasis, and evasion of immune destruction, and discuss how collectively these hallmarks could be tackled by therapies targeting pericytes, providing a rationale for cancer drugs aiming at the TME.
Collapse
|
123
|
Endogenous brain pericytes are widely activated and contribute to mouse glioma microvasculature. PLoS One 2015; 10:e0123553. [PMID: 25875288 PMCID: PMC4395339 DOI: 10.1371/journal.pone.0123553] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/05/2015] [Indexed: 02/08/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common brain tumor in adults. It presents an extremely challenging clinical problem, and treatment very frequently fails due to the infiltrative growth, facilitated by extensive angiogenesis and neovascularization. Pericytes constitute an important part of the GBM microvasculature. The contribution of endogenous brain pericytes to the tumor vasculature in GBM is, however, unclear. In this study, we determine the site of activation and the extent of contribution of endogenous brain pericytes to the GBM vasculature. GL261 mouse glioma was orthotopically implanted in mice expressing green fluorescent protein (GFP) under the pericyte marker regulator of G protein signaling 5 (RGS5). Host pericytes were not only activated within the glioma, but also in cortical areas overlying the tumor, the ipsilateral subventricular zone and within the hemisphere contralateral to the tumor. The host-derived activated pericytes that infiltrated the glioma were mainly localized to the tumor vessel wall. Infiltrating GFP positive pericytes co-expressed the pericyte markers platelet-derived growth factor receptor-β (PDGFR-β) and neuron-glial antigen 2. Interestingly, more than half of all PDGFR-β positive pericytes within the tumor were contributed by the host brain. We did not find any evidence that RGS5 positive pericytes adopt another phenotype within glioma in this paradigm. We conclude that endogenous pericytes become activated in widespread areas of the brain in response to an orthotopic mouse glioma. Host pericytes are recruited into the tumor and constitute a major part of the tumor pericyte population.
Collapse
|
124
|
The cholesterol biosynthesis enzyme oxidosqualene cyclase is a new target to impair tumour angiogenesis and metastasis dissemination. Sci Rep 2015; 5:9054. [PMID: 25761781 PMCID: PMC4357009 DOI: 10.1038/srep09054] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 02/16/2015] [Indexed: 12/13/2022] Open
Abstract
Aberrant cholesterol homeostasis and biosynthesis has been observed in different tumour types. This paper investigates the role of the post-squalenic enzyme of cholesterol biosynthesis, oxidosqualene cyclase (OSC), in regulating tumour angiogenesis and metastasis dissemination in mouse models of cancer. We showed that Ro 48-8071, a selective inhibitor of OSC, reduced vascular density and increased pericyte coverage, with a consequent inhibition of tumour growth in a spontaneous mouse model of pancreatic tumour (RIP-Tag2) and two metastatic mouse models of human colon carcinoma (HCT116) and pancreatic adenocarcinoma (HPAF-II). Remarkably, the inhibition of OSC hampered metastasis formation in HCT116 and HPAF-II models. Ro 48-8071 induced tumour vessel normalization and enhanced the anti-tumoral and anti-metastatic effects of 5-fluorouracil (5-FU) in HCT116 mice. Ro 48-8071 exerted a strong anti-angiogenic activity by impairing endothelial cell adhesion and migration, and by blocking vessel formation in angiogenesis assays. OSC inhibition specifically interfered with the PI3K pathway. According to in vitro results, Ro 48-8071 specifically inhibited Akt phosphorylation in both cancer cells and tumour vasculature in all treated models. Thus, our results unveil a crucial role of OSC in the regulation of cancer progression and tumour angiogenesis, and indicate Ro 48-8071 as a potential novel anti-angiogenic and anti-metastatic drug.
Collapse
|
125
|
Rivera LB, Bergers G. Intertwined regulation of angiogenesis and immunity by myeloid cells. Trends Immunol 2015; 36:240-9. [PMID: 25770923 DOI: 10.1016/j.it.2015.02.005] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/16/2015] [Accepted: 02/17/2015] [Indexed: 12/11/2022]
Abstract
Angiogenesis is a hallmark of cancer because its induction is indispensable to fuel an expanding tumor. The tumor microenvironment contributes to tumor vessel growth, and distinct myeloid cells recruited by the tumor have been shown not only to support angiogenesis but also to foster an immune suppressive environment that supports tumor expansion and progression. Recent findings suggest that the intertwined regulation of angiogenesis and immune modulation can offer therapeutic opportunities for the treatment of cancer. We review the mechanisms by which distinct myeloid cell populations contribute to tumor angiogenesis, discuss current approaches in the clinic that are targeting both angiogenic and immune suppressive pathways, and highlight important areas of future research.
Collapse
Affiliation(s)
- Lee B Rivera
- Department of Neurological Surgery, University of California, San Francisco, CA 94158, USA; University of California San Francisco (UCSF) Comprehensive Cancer Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, CA 94158, USA.
| | - Gabriele Bergers
- Department of Neurological Surgery, University of California, San Francisco, CA 94158, USA; University of California San Francisco (UCSF) Comprehensive Cancer Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, CA 94158, USA; Brain Tumor Center, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
126
|
Heindryckx F, Gerwins P. Targeting the tumor stroma in hepatocellular carcinoma. World J Hepatol 2015; 7:165-176. [PMID: 25729472 PMCID: PMC4342599 DOI: 10.4254/wjh.v7.i2.165] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/30/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common and deadly cancers worldwide. In ninety percent of the cases it develops as a result of chronic liver damage and it is thus a typical inflammation-related cancer characterized by the close relation between the tumor microenvironment and tumor cells. The stromal environment consists out of several cell types, including hepatic stellate cells, macrophages and endothelial cells. They are not just active bystanders in the pathogenesis of HCC, but play an important and active role in tumor initiation, progression and metastasis. Furthermore, the tumor itself influences these cells to create a background that is beneficial for sustaining tumor growth. One of the key players is the hepatic stellate cell, which is activated during liver damage and differentiates towards a myofibroblast-like cell. Activated stellate cells are responsible for the deposition of extracellular matrix, increase the production of angiogenic factors and stimulate the recruitment of macrophages. The increase of angiogenic factors (which are secreted by macrophages, tumor cells and activated stellate cells) will induce the formation of new blood vessels, thereby supplying the tumor with more oxygen and nutrients, thus supporting tumor growth and offering a passageway in the circulatory system. In addition, the secretion of chemokines by the tumor cells leads to the recruitment of tumor associated macrophages. These tumor associated macrophages are key actors of cancer-related inflammation, being the main type of inflammatory cells infiltrating the tumor environment and exerting a tumor promoting effect by secreting growth factors, stimulating angiogenesis and influencing the activation of stellate cells. This complex interplay between the several cell types involved in liver cancer emphasizes the need for targeting the tumor stroma in HCC patients.
Collapse
|
127
|
Future therapies of wet age-related macular degeneration. J Ophthalmol 2015; 2015:138070. [PMID: 25802751 PMCID: PMC4354726 DOI: 10.1155/2015/138070] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/10/2015] [Indexed: 01/08/2023] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness in the elderly population, and the prevalence of the disease increases exponentially with every decade after the age of 50 years. While VEGF inhibitors are promising drugs for treating patients with ocular neovascularization, there are limitations to their potential for improving vision in AMD patients. Thus, future therapies are required to have the potential to improve visual outcomes. This paper will summarize the future strategies and therapeutic targets that are aimed at enhancing the efficacy and duration of effect of antiangiogenic strategies.
Collapse
|
128
|
Keskin D, Kim J, Cooke VG, Wu CC, Sugimoto H, Gu C, De Palma M, Kalluri R, LeBleu VS. Targeting vascular pericytes in hypoxic tumors increases lung metastasis via angiopoietin-2. Cell Rep 2015; 10:1066-81. [PMID: 25704811 DOI: 10.1016/j.celrep.2015.01.035] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 12/02/2014] [Accepted: 01/13/2015] [Indexed: 01/12/2023] Open
Abstract
Strategies to target angiogenesis include inhibition of the vessel-stabilizing properties of vascular pericytes. Pericyte depletion in early-stage non-hypoxic tumors suppressed nascent angiogenesis, tumor growth, and lung metastasis. In contrast, pericyte depletion in advanced-stage hypoxic tumors with pre-established vasculature resulted in enhanced intra-tumoral hypoxia, decreased tumor growth, and increased lung metastasis. Furthermore, depletion of pericytes in post-natal retinal blood vessels resulted in abnormal and leaky vasculature. Tumor transcriptome profiling and biological validation revealed that angiopoietin signaling is a key regulatory pathway associated with pericyte targeting. Indeed, pericyte targeting in established mouse tumors increased angiopoietin-2 (ANG2/Angpt2) expression. Depletion of pericytes, coupled with targeting of ANG2 signaling, restored vascular stability in multiple model systems and decreased tumor growth and metastasis. Importantly, ANGPT2 expression correlated with poor outcome in patients with breast cancer. These results emphasize the potential utility of therapeutic regimens that target pericytes and ANG2 signaling in metastatic breast cancer.
Collapse
Affiliation(s)
- Doruk Keskin
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Jiha Kim
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Vesselina G Cooke
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Chia-Chin Wu
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Hikaru Sugimoto
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Michele De Palma
- The Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA.
| | - Valerie S LeBleu
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
129
|
Kanada M, Zhang J, Yan L, Sakurai T, Terakawa S. Endothelial cell-initiated extravasation of cancer cells visualized in zebrafish. PeerJ 2014; 2:e688. [PMID: 25551022 PMCID: PMC4277486 DOI: 10.7717/peerj.688] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 11/17/2014] [Indexed: 12/15/2022] Open
Abstract
The extravasation of cancer cells, a key step for distant metastasis, is thought to be initiated by disruption of the endothelial barrier by malignant cancer cells. An endothelial covering-type extravasation of cancer cells in addition to conventional cancer cell invasion-type extravasation was dynamically visualized in a zebrafish hematogenous metastasis model. The inhibition of VEGF-signaling impaired the invasion-type extravasation via inhibition of cancer cell polarization and motility. Paradoxically, the anti-angiogenic treatment showed the promotion, rather than the inhibition, of the endothelial covering-type extravasation of cancer cells, with structural changes in the endothelial walls. These findings may be a set of clues to the full understanding of the metastatic process as well as the metastatic acceleration by anti-angiogenic reagents observed in preclinical studies.
Collapse
Affiliation(s)
- Masamitsu Kanada
- Medical Photonics Research Center, Hamamatsu University School of Medicine , Handayama, Higashi-ku, Hamamatsu , Japan
| | - Jinyan Zhang
- Medical Photonics Research Center, Hamamatsu University School of Medicine , Handayama, Higashi-ku, Hamamatsu , Japan
| | - Libo Yan
- Medical Photonics Research Center, Hamamatsu University School of Medicine , Handayama, Higashi-ku, Hamamatsu , Japan
| | - Takashi Sakurai
- Electronics-inspired Interdiciplinary Research Institute, Toyohashi University of Technology , Hibarigaoka, Tempaku, Toyohashi , Japan
| | - Susumu Terakawa
- Medical Photonics Research Center, Hamamatsu University School of Medicine , Handayama, Higashi-ku, Hamamatsu , Japan
| |
Collapse
|
130
|
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: 1.5] [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
|
131
|
Tudoran OM, Soritau O, Balacescu L, Pop L, Meurice G, Visan S, Lindberg S, Eniu A, Langel U, Balacescu O, Berindan-Neagoe I. PDGF beta targeting in cervical cancer cells suggest a fine-tuning of compensatory signalling pathways to sustain tumourigenic stimulation. J Cell Mol Med 2014; 19:371-82. [PMID: 25311137 PMCID: PMC4407607 DOI: 10.1111/jcmm.12449] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 08/25/2014] [Indexed: 12/18/2022] Open
Abstract
The platelet-derived growth factor (PDGF) signalling pathway has been reported to play an important role in human cancers by modulating autocrine and paracrine processes such as tumour growth, metastasis and angiogenesis. Several clinical trials document the benefits of targeting this pathway; however, in cervical cancer the role of PDGF signalling in still unclear. In this study, we used siRNA against PDGF beta (PDGFBB) to investigate the cellular and molecular mechanisms of PDGFBB signalling in Ca Ski and HeLa cervical cancer cells. Our results show that PDGFBB inhibition in Ca Ski cells led to rapid alterations of the transcriptional pattern of 579 genes, genes that are known to have antagonistic roles in regulating tumour progression. Concomitantly, with the lack of significant effects on cervical cancer cells proliferation, apoptosis, migration or invasion, these findings suggests that cervical cancer cells shift between compensatory signalling pathways to maintain their behaviour. The observed autocrine effects were limited to cervical cancer cells ability to adhere to an endothelial cell (EC) monolayer. However, by inhibiting PDGFBB on cervical cells, we achieved reduced proliferation of ECs in co-culture settings and cellular aggregation in conditioned media. Because of lack of PDGF receptor expression on ECs, we believe that these effects are a result of indirect PDGFBB paracrine signalling mechanisms. Our results shed some light into the understanding of PDGFBB signalling mechanism in cervical cancer cells, which could be further exploited for the development of synergistic anti-tumour and anti-angiogenic therapeutic strategies.
Collapse
Affiliation(s)
- Oana Mihaela Tudoran
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. I. Chiricuta", Cluj-Napoca, Romania
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Pinto MP, Dye WW, Jacobsen BM, Horwitz KB. Malignant stroma increases luminal breast cancer cell proliferation and angiogenesis through platelet-derived growth factor signaling. BMC Cancer 2014; 14:735. [PMID: 25274034 PMCID: PMC4190420 DOI: 10.1186/1471-2407-14-735] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 09/26/2014] [Indexed: 12/21/2022] Open
Abstract
Background Luminal, estrogen receptor-positive breast cancers represent more than 70% of cases. Despite initial good prognoses one third of Luminal cancers eventually recur locally or at distant sites and exhibit hormone resistance. Here we demonstrate that factors elaborated by malignant stromal cells can induce Luminal tumor cells proliferation and promote angiogenesis and hormone independence. We recently isolated a malignant mouse mammary gland stromal cell line named BJ3Z that increases proliferation and angiogenesis in estrogen-free xenografted Luminal MCF-7 breast cancer cells. Methods BJ3Z and Normal mouse mammary Fibroblasts (NMFs) were expression profiled using microarray assays. Messenger RNA levels were confirmed by RT-PCR and by immunohistochemistry (IHC). Breast cancer MCF-7, BT-474, BT-20 and MDA-MB-231cell lines and stromal BJ3Z and NMFs were grown for in vitro assays: breast cancer cell lines were treated with stromal cells conditioned media, for three-dimensional (3D) mono and co-cultures in Matrigel, proliferation was measured by Bromo-deoxyuridine (BrdU) incorporation using IHC. Tubule formation in vitro, a proxy for angiogenesis, was assessed using 3D cultured Human Umbilical cord Vascular Endothelial Cells (HUVEC). Results We show that under estrogen-free conditions, BJ3Z cells but not NMFs increase proliferation of co-cultured Luminal but not basal-like human breast cancer cells in 2D or as 3D Matrigel colonies. Gene expression profiling, RT-PCR analysis and IHC of colony-derived BJ3Z cells and NMFs shows that Platelet Derived Growth Factor ligands (PDGF-A and -B) are elaborated by BJ3Z cells but not NMFs; while PDGF receptors are present on NMFs but not BJ3Z cells. As a result, in colony co-culture assays, BJ3Z cells but not NMFs increase MCF-7 cell proliferation. This can be mimicked by direct addition of PDGF-BB, and blocked by the PDGF receptor inhibitor Imatinib Mesylate. Both normal and malignant stromal cells enhance angiogenesis in an in vitro model. This effect is also due to PDGF and is suppressed by Imatinib. Conclusions We provide evidence that Luminal breast cancer cells can be targeted by the PDGF signaling pathway leading to estrogen-independent proliferation and angiogenesis. We speculate that stroma-directed therapies, including anti-PDGFR agents like Imatinib, may be useful in combination with other therapies for treatment of luminal cancers. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-735) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mauricio P Pinto
- Departments of Medicine, Mail Stop 8106, 12801 East 17th Avenue, Aurora, CO 80045, USA.
| | | | | | | |
Collapse
|
133
|
The inhibition of tyrosine kinase receptor signalling in leiomyosarcoma cells using the small molecule kinase inhibitor PTK787/ZK222584 (Vatalanib®). Int J Oncol 2014; 45:2267-77. [PMID: 25340839 PMCID: PMC4215578 DOI: 10.3892/ijo.2014.2683] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/03/2014] [Indexed: 12/21/2022] Open
Abstract
Leiomyosarcomas remain challenging tumors to manage and novel therapy strategies besides radiation and conventional chemotherapy are needed. Targeting angiogenesis by inhibition of vascular endothelial growth factor (VEGF) receptor tyrosine kinases (RTKs) of the tumor vasculature with small molecules is a promising new therapy. It has been shown recently that these receptors are not only expressed on tumor endothelium but also on tumor cells themselves. Thus, we investigated the expression of members of the VEGF receptor (VEGFR) family and corresponding growth factors in leiomyosarcoma tissue specimens and in the leiomyosarcoma cell lines SK-LMS-1 and SK-UT-1. We evaluated the influence of the VEGFR inhibitor PTK787/ZK222584 (PTK787) on cell growth, migration, apoptosis and phosphorylation of intracellular signalling molecules. In human leiomyosarcoma tissue specimens VEGFR‑1/-2 and platelet-derived growth factor receptor (PDGFR-β) were strongly expressed. Both leiomyosarcoma cell lines expressed VEGFR‑1/-3 and PDGFR-β but VEGFR-2 protein expression was positive only in SK-UT-1. SK-LMS-1 and SK-UT-1 cells secreted high and low amounts of VEGF-A, respectively, whereas PDGF-BB secretion was similar in both cell lines. Application of PTK787 led to partial inhibition of PDGF-BB-activated AKT/p90RSK and ERK1/2 signalling pathways. In contrast, protein phosphorylation was not affected by PTK787 in VEGF-A-treated cells. PTK787 turned out to inhibit cell migration even though no effects were observed upon stimulation with VEGF-A or PDGF-BB. In line, cell growth in leiomyosarcoma cell lines remained unchanged upon PTK787 treatment alone and with subsequent VEGF-A- or PDGF-BB-stimulation. However, VEGF-A, but not PDGF-BB-treated cells showed increased cell death upon PTK787 treatment. VEGFR family members are expressed in leiomyosarcomas in vivo and in vitro. Upon receptor stimulation, PTK787 is able to inhibit subsequent phosphorylation events and influences cell survival but not metabolic activity and migration. Thus, the inhibitor is possibly an additional option in the treatment of leiomyosarcomas.
Collapse
|
134
|
Combining molecular targeted drugs to inhibit both cancer cells and activated stromal cells in gastric cancer. Neoplasia 2014; 15:1391-9. [PMID: 24403861 DOI: 10.1593/neo.131668] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 09/20/2013] [Accepted: 11/06/2013] [Indexed: 11/18/2022] Open
Abstract
Recent studies have revealed that PDGF plays a role in promoting progressive tumor growth in several cancers, including gastric cancer. Cancer-associated fibroblasts, pericytes, and lymphatic endothelial cells in stroma express high levels of PDGF receptor (PDGF-R); cancer cells and vascular endothelial cells do not. Mammalian target of rapamycin (mTOR) is a serine/threonine kinase that increases the production of proteins that stimulate key cellular processes such as cell growth and proliferation, cell metabolism, and angiogenesis. In the present study, we examined the effects of PDGF-R tyrosine kinase inhibitor (nilotinib) and mTOR inhibitor (everolimus) on tumor stroma in an orthotopic nude mice model of human gastric cancer. Expression of PDGF-B and PDGF-Rβ mRNAs was associated with stromal volume. Treatment with nilotinib did not suppress tumor growth but significantly decreased stromal reactivity, lymphatic invasion, lymphatic vessel area, and pericyte coverage of tumor microvessels. In contrast, treatment with everolimus decreased tumor growth and microvessel density but not stromal reactivity. Nilotinib and everolimus in combination reduced both the growth rate and stromal reaction. Target molecule-based inhibition of cancer-stromal cell interaction appears promising as an effective antitumor therapy.
Collapse
|
135
|
Rothschild SI. Clinical potential of nintedanib for the second-line treatment of advanced non-small-cell lung cancer: current evidence. LUNG CANCER-TARGETS AND THERAPY 2014; 5:51-57. [PMID: 28210142 PMCID: PMC5217508 DOI: 10.2147/lctt.s49490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The therapeutic landscape in non-small-cell lung cancer (NSCLC) is changing. The description of molecular alterations leading to NSCLC carcinogenesis and progression (so-called oncogenic driver mutations) and the development of targeted agents interfering with the tumor-promoting intracellular signaling pathways have improved the outcome for many patients with advanced/metastatic NSCLC. However, many patients with stage IV NSCLC do not have one of the targetable predictive biomarkers, and are therefore in need of classical chemotherapy. This especially applies to squamous cell cancer. A platinum-based doublet chemotherapy is the standard of care for patients with stage IV NSCLC. As second-line therapies, docetaxel, pemetrexed, and the EGFR tyrosine-kinase inhibitor erlotinib have demonstrated benefit in Phase III randomized trials. Recently, the addition of the angiokinase inhibitor nintedanib to docetaxel has proven efficacious, and is a new treatment option in the second-line setting. Preclinical and clinical data of nintedanib for the treatment of lung cancer patients are reviewed here.
Collapse
Affiliation(s)
- Sacha I Rothschild
- Department of Internal Medicine, Medical Oncology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
136
|
Backen A, Renehan AG, Clamp AR, Berzuini C, Zhou C, Oza A, Bannoo S, Scherer SJ, Banks RE, Dive C, Jayson GC. The combination of circulating Ang1 and Tie2 levels predicts progression-free survival advantage in bevacizumab-treated patients with ovarian cancer. Clin Cancer Res 2014; 20:4549-4558. [PMID: 24947924 PMCID: PMC4154862 DOI: 10.1158/1078-0432.ccr-13-3248] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE Randomized ovarian cancer trials, including ICON7, have reported improved progression-free survival (PFS) when bevacizumab was added to conventional cytotoxic therapy. The improvement was modest prompting the search for predictive biomarkers for bevacizumab. EXPERIMENTAL DESIGN Pretreatment training (n=91) and validation (n=114) blood samples were provided by ICON7 patients. Plasma concentrations of 15 angio-associated factors were determined using validated multiplex ELISAs. Our statistical approach adopted PFS as the primary outcome measure and involved (i) searching for biomarkers with prognostic relevance or which related to between-individual variation in bevacizumab effect; (ii) unbiased determination of cutoffs for putative biomarker values; (iii) investigation of biologically meaningfully predictive combinations of putative biomarkers; and (iv) replicating the analysis on candidate biomarkers in the validation dataset. RESULTS The combined values of circulating Ang1 (angiopoietin 1) and Tie2 (Tunica internal endothelial cell kinase 2) concentrations predicted improved PFS in bevacizumab-treated patients in the training set. Using median concentrations as cutoffs, high Ang1/low Tie2 values were associated with significantly improved PFS for bevacizumab-treated patients in both datasets (median, 23.0 months vs. 16.2; P=0.003) for the interaction of Ang1-Tie2 treatment in Cox regression analysis. The prognostic indices derived from the training set also distinguished high and low probability for progression in the validation set (P=0.008), generating similar values for HR (0.21 vs. 0.27) between treatment and control arms for patients with high Ang1 and low Tie2 values. CONCLUSIONS The combined values of Ang1 and Tie2 are predictive biomarkers for improved PFS in bevacizumab-treated patients with ovarian cancer. These findings need to be validated in larger trials due to the limitation of sample size in this study.
Collapse
Affiliation(s)
- Alison Backen
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester M20 4BX, United Kingdom
| | - Andrew G Renehan
- Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Center, The Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| | - Andrew R Clamp
- Dept Medical Oncology, University of Manchester and Christie Hospital, Wilmslow Road, Withington, Manchester M20 4BX, UK
| | - Carlo Berzuini
- Center for Biostatistics, Institute of Population Health, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Cong Zhou
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester M20 4BX, United Kingdom
| | - Amit Oza
- Bras Family Drug Development Program, Princess Margaret Hospital, 610 University Avenue, Toronto M5G 2M9
| | - Selina Bannoo
- MRC Clinical Trials Unit, Aviation House, 125 Kingsway, London WC2B 6NH
| | - Stefan J Scherer
- Biocartis SA, EPFL-Quartier de l'Innovation, Bâtimont G, CH-1015 Lausanne
| | - Rosamonde E Banks
- Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds LS9 7TF, UK
| | - Caroline Dive
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester M20 4BX, United Kingdom
| | - Gordon C Jayson
- Translational Angiogenesis Group, University of Manchester and Christie Hospital, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| |
Collapse
|
137
|
Backen A, Renehan AG, Clamp AR, Berzuini C, Zhou C, Oza A, Bannoo S, Scherer SJ, Banks RE, Dive C, Jayson GC. The combination of circulating Ang1 and Tie2 levels predicts progression-free survival advantage in bevacizumab-treated patients with ovarian cancer. Clin Cancer Res 2014. [PMID: 24947924 DOI: 10.1158/1078-0432.ccr-13-3248] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Randomized ovarian cancer trials, including ICON7, have reported improved progression-free survival (PFS) when bevacizumab was added to conventional cytotoxic therapy. The improvement was modest prompting the search for predictive biomarkers for bevacizumab. EXPERIMENTAL DESIGN Pretreatment training (n=91) and validation (n=114) blood samples were provided by ICON7 patients. Plasma concentrations of 15 angio-associated factors were determined using validated multiplex ELISAs. Our statistical approach adopted PFS as the primary outcome measure and involved (i) searching for biomarkers with prognostic relevance or which related to between-individual variation in bevacizumab effect; (ii) unbiased determination of cutoffs for putative biomarker values; (iii) investigation of biologically meaningfully predictive combinations of putative biomarkers; and (iv) replicating the analysis on candidate biomarkers in the validation dataset. RESULTS The combined values of circulating Ang1 (angiopoietin 1) and Tie2 (Tunica internal endothelial cell kinase 2) concentrations predicted improved PFS in bevacizumab-treated patients in the training set. Using median concentrations as cutoffs, high Ang1/low Tie2 values were associated with significantly improved PFS for bevacizumab-treated patients in both datasets (median, 23.0 months vs. 16.2; P=0.003) for the interaction of Ang1-Tie2 treatment in Cox regression analysis. The prognostic indices derived from the training set also distinguished high and low probability for progression in the validation set (P=0.008), generating similar values for HR (0.21 vs. 0.27) between treatment and control arms for patients with high Ang1 and low Tie2 values. CONCLUSIONS The combined values of Ang1 and Tie2 are predictive biomarkers for improved PFS in bevacizumab-treated patients with ovarian cancer. These findings need to be validated in larger trials due to the limitation of sample size in this study.
Collapse
Affiliation(s)
- Alison Backen
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester M20 4BX, United Kingdom
| | - Andrew G Renehan
- Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Center, The Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| | - Andrew R Clamp
- Dept Medical Oncology, University of Manchester and Christie Hospital, Wilmslow Road, Withington, Manchester M20 4BX, UK
| | - Carlo Berzuini
- Center for Biostatistics, Institute of Population Health, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Cong Zhou
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester M20 4BX, United Kingdom
| | - Amit Oza
- Bras Family Drug Development Program, Princess Margaret Hospital, 610 University Avenue, Toronto M5G 2M9
| | - Selina Bannoo
- MRC Clinical Trials Unit, Aviation House, 125 Kingsway, London WC2B 6NH
| | - Stefan J Scherer
- Biocartis SA, EPFL-Quartier de l'Innovation, Bâtimont G, CH-1015 Lausanne
| | - Rosamonde E Banks
- Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds LS9 7TF, UK
| | - Caroline Dive
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester M20 4BX, United Kingdom
| | - Gordon C Jayson
- Translational Angiogenesis Group, University of Manchester and Christie Hospital, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| |
Collapse
|
138
|
Mross K, Büchert M, Frost A, Medinger M, Stopfer P, Studeny M, Kaiser R. Vascular effects, efficacy and safety of nintedanib in patients with advanced, refractory colorectal cancer: a prospective phase I subanalysis. BMC Cancer 2014; 14:510. [PMID: 25012508 PMCID: PMC4105047 DOI: 10.1186/1471-2407-14-510] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 07/04/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Nintedanib is a potent, oral angiokinase inhibitor that targets VEGF, PDGF and FGF signalling, as well as RET and Flt3. The maximum tolerated dose of nintedanib was evaluated in a phase I study of treatment-refractory patients with advanced solid tumours. In this preplanned subanalysis, the effect of nintedanib on the tumour vasculature, along with efficacy and safety, was assessed in 30 patients with colorectal cancer (CRC). METHODS Patients with advanced CRC who had failed conventional treatment, or for whom no therapy of proven efficacy existed, were treated with nintedanib ranging from 50-450 mg once-daily (n = 14) or 150-250 mg twice-daily (n = 16) for 28 days. After a 1-week rest, further courses were permitted in the absence of progression or undue toxicity. The primary objective was the effect on the tumour vasculature using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and expressed as the initial area under the DCE-MRI contrast agent concentration-time curve after 60 seconds (iAUC60) or the volume transfer constant between blood plasma and extravascular extracellular space (Ktrans). RESULTS Patients received a median of 4.0 courses (range: 1-13). Among 21 evaluable patients, 14 (67%) had a ≥40% reduction from baseline in Ktrans and 13 (62%) had a ≥40% decrease from baseline in iAUC60, representing clinically relevant effects on tumour blood flow and permeability, respectively. A ≥40% reduction from baseline in Ktrans was positively associated with non-progressive tumour status (Fisher's exact: p = 0.0032). One patient achieved a partial response at 250 mg twice-daily and 24 (80%) achieved stable disease lasting ≥8 weeks. Time to tumour progression (TTP) at 4 months was 26% and median TTP was 72.5 days (95% confidence interval: 65-114). Common drug-related adverse events (AEs) included nausea (67%), vomiting (53%) and diarrhoea (40%); three patients experienced drug-related AEs ≥ grade 3. Four patients treated with nintedanib once-daily had an alanine aminotransferase and/or aspartate aminotransferase increase ≥ grade 3. No increases > grade 2 were seen in the twice-daily group. CONCLUSIONS Nintedanib modulates tumour blood flow and permeability in patients with advanced, refractory CRC, while achieving antitumour activity and maintaining an acceptable safety profile.
Collapse
Affiliation(s)
- Klaus Mross
- Tumor Biology Center, Department of Medical Oncology, Breisacherstrasse 117, D-79106 Freiburg in Breisgau, Germany
| | - Martin Büchert
- Magnetic Resonance Development and Application Center, Department of Radiology, University Medical Center Freiburg, Freiburg, Germany
| | - Annette Frost
- Tumor Biology Center, Department of Medical Oncology, Breisacherstrasse 117, D-79106 Freiburg in Breisgau, Germany
| | - Michael Medinger
- Tumor Biology Center, Department of Medical Oncology, Breisacherstrasse 117, D-79106 Freiburg in Breisgau, Germany
| | - Peter Stopfer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Matus Studeny
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Rolf Kaiser
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
139
|
Zhao XY, Yang S, Chen YR, Li PC, Dou MM, Zhang J. Resveratrol and arsenic trioxide act synergistically to kill tumor cells in vitro and in vivo. PLoS One 2014; 9:e98925. [PMID: 24901647 PMCID: PMC4047048 DOI: 10.1371/journal.pone.0098925] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 05/08/2014] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND AIMS Arsenic trioxide (As2O3), which used as an effective agent in the treatment of leukaemia and other solid tumors, is largely limited by its toxicity. QT prolongation, torsades de pointes and sudden heart death have been implicated in the cardiotoxicity of As2O3. The present study was designed to explore whether the combination of As2O3 and resveratrol could generate a more powerful anti-cancer effect both in vitro and in vivo. MATERIALS AND METHODS MTT assay was performed to assess the proliferation of Hela, MCF-7 and NB4 cells. Isobolographic analysis was used to evaluate combination index values from cell viability data. The apoptosis and the cellular reactive oxygen species (ROS) level were assessed by fluorescent microscopy and flow cytometry separately in vitro. The effect of As2O3, alone and in combination with resveratrol on Hela tumor growth in an orthotopic nude mouse model was also investigated. The tumor volume and the immunohistochemical analysis of CD31, CD34 and VEGF were determined. RESULTS Resveratrol dramatically enhanced the anti-cancer effect induced by As2O3 in vitro. In addition, isobolographic analysis further demonstrated that As2O3 and resveratrol generated a synergistic action. More apoptosis and ROS generation were observed in the combination treatment group. Similar synergistic effects were found in nude mice in vivo. The combination of As2O3 and resveratrol dramatically suppressed both tumor growth and angiogenesis in nude mice. CONCLUSIONS Combining As2O3 with resveratrol would be a novel strategy to treat cancer in clinical practice.
Collapse
Affiliation(s)
- Xiao-Yan Zhao
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
- * E-mail: (JZ); (XZ)
| | - Shen Yang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - You-Ran Chen
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Pei-Chun Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Meng-Meng Dou
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Jie Zhang
- Department of Neurology, The Ninth People’s Hospital of Chongqing, Chongqing, China
- * E-mail: (JZ); (XZ)
| |
Collapse
|
140
|
Stapor PC, Sweat RS, Dashti DC, Betancourt AM, Murfee WL. Pericyte dynamics during angiogenesis: new insights from new identities. J Vasc Res 2014; 51:163-74. [PMID: 24853910 DOI: 10.1159/000362276] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 03/11/2014] [Indexed: 12/13/2022] Open
Abstract
Therapies aimed at manipulating the microcirculation require the ability to control angiogenesis, defined as the sprouting of new capillaries from existing vessels. Blocking angiogenesis would be beneficial in many pathologies (e.g. cancer, retinopathies and rheumatoid arthritis). In others (e.g. myocardial infarction, stroke and hypertension), promoting angiogenesis would be desirable. We know that vascular pericytes elongate around endothelial cells (ECs) and are functionally associated with regulating vessel stabilization, vessel diameter and EC proliferation. During angiogenesis, bidirectional pericyte-EC signaling is critical for capillary sprout formation. Observations of pericytes leading capillary sprouts also implicate their role in EC guidance. As such, pericytes have recently emerged as a therapeutic target to promote or inhibit angiogenesis. Advancing our basic understanding of pericytes and developing pericyte-related therapies are challenged, like in many other fields, by questions regarding cell identity. This review article discusses what we know about pericyte phenotypes and the opportunity to advance our understanding by defining the specific pericyte cell populations involved in capillary sprouting.
Collapse
Affiliation(s)
- Peter C Stapor
- Department of Biomedical Engineering, Tulane University, Lindy Boggs Center, New Orleans, La., USA
| | | | | | | | | |
Collapse
|
141
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Olson JD, Mintz A, Delbono O. Type-2 pericytes participate in normal and tumoral angiogenesis. Am J Physiol Cell Physiol 2014; 307:C25-38. [PMID: 24788248 DOI: 10.1152/ajpcell.00084.2014] [Citation(s) in RCA: 230] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tissue growth and function depend on vascularization, and vascular insufficiency or excess exacerbates many human diseases. Identification of the biological processes involved in angiogenesis will dictate strategies to modulate reduced or excessive vessel formation. We examine the essential role of pericytes. Their heterogeneous morphology, distribution, origins, and physiology have been described. Using double-transgenic Nestin-GFP/NG2-DsRed mice, we identified two pericyte subsets. We found that Nestin-GFP(-)/NG2-DsRed(+) (type-1) and Nestin-GFP(+)/NG2-DsRed(+) (type-2) pericytes attach to the walls of small and large blood vessels in vivo; in vitro, type-2, but not type-1, pericytes spark endothelial cells to form new vessels. Matrigel assay showed that only type-2 pericytes participate in normal angiogenesis. Moreover, when cancer cells were transplanted into Nestin-GFP/NG2-DsRed mice, type-1 pericytes did not penetrate the tumor, while type-2 pericytes were recruited during its angiogenesis. As inhibition of angiogenesis is a promising strategy in cancer therapy, type-2 pericytes may provide a cellular target susceptible to signaling and pharmacological manipulation in treating malignancy. This work also reports the potential of type-2 pericytes to improve blood perfusion in ischemic hindlimbs, indicating their potential for treating ischemic illnesses.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina; Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Maria Laura Messi
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - John D Olson
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina; and
| | - Akiva Mintz
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina; and Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina; Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina;
| |
Collapse
|
142
|
Tian LL, Ren B, Gao XW, Luo Y, Cai Y, Zhou K, Du AJ, Zhao Y. Inhibition of retinopathy of prematurity in rat by intravitreal injection of sorafenib. Int J Ophthalmol 2014; 7:198-204. [PMID: 24790858 DOI: 10.3980/j.issn.2222-3959.2014.02.03] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/11/2013] [Indexed: 12/23/2022] Open
Abstract
AIM To investigate the effect of intravitreal injection administered sorafenib, a multikinase inhibitor, in a rat model of oxygen-induced retinopathy (OIR). METHODS Seven-day-old Sprague-Dawley rats (n=144) were randomly assigned to six groups. Group A received normal partial oxygen pressure and groups B, C, D, E and F were exposed to hyperoxia (75±2)% from postnatal 7d (P7) to P12 to induce retinopathy of prematurity. The rats in groups C, D, E and F were received intravitreal injections of either vehicle (DMSO) or sorafenib at P12 (5, 20 and 80 µg, respectively). Then they returned to normoxia after P12. The retinas were whole-mounted and imaged with a confocal microscopy. The vascular branching points were counted to quantify neovascularization at P17. Cross-sections of the retina were stained with hematoxylin and eosin (HE). The nuclei of new vessels breaking the internal limiting membrane were counted to quantify the proliferative neovascular response. RESULTS The retinal vessel in groups B and C turned into tortuosity and a great deal of neovascularization were observed. Sorafenib-treated rats had significantly less neovascularization as compared with vehicle-treated and control rats in a dose dependent manner (P<0.05). The number of vascular branching points in A, B, C, D, E and F were 16.50±3.90, 37.44±6.47, 37.08±5.10, 30.80±6.85, 26.08±5.08 and 19.83±3.51, respectively. The number of the nuclei of retinal new vessel in A, B, C, D, E and F were 0.22±0.42, 35.66±4.70, 35.30±4.54, 27.30±4.28, 21.41±3.53, and 7.41±2.87, respectively. There were significant difference between each group (P<0.05) except groups B and C. CONCLUSION In the rat OIR model, sorafenib could inhibit retinal neovascularization in a dose dependent manner.
Collapse
Affiliation(s)
- Li-Li Tian
- Medical College of Shihezi University, Shihezi 832000, Xinjiang Uygur Autonomous Region, China ; Ophthalmic Center, No.474 Hospital of Chinese PLA, Urumqi 830013, Xinjiang Uygur Autonomous Region, China
| | - Bing Ren
- Ophthalmic Center, No.474 Hospital of Chinese PLA, Urumqi 830013, Xinjiang Uygur Autonomous Region, China
| | - Xiao-Wei Gao
- Ophthalmic Center, No.474 Hospital of Chinese PLA, Urumqi 830013, Xinjiang Uygur Autonomous Region, China
| | - Ying Luo
- Ophthalmic Center, No.474 Hospital of Chinese PLA, Urumqi 830013, Xinjiang Uygur Autonomous Region, China
| | - Yan Cai
- Ophthalmic Center, No.474 Hospital of Chinese PLA, Urumqi 830013, Xinjiang Uygur Autonomous Region, China
| | - Kun Zhou
- Medical College of Shihezi University, Shihezi 832000, Xinjiang Uygur Autonomous Region, China ; Ophthalmic Center, No.474 Hospital of Chinese PLA, Urumqi 830013, Xinjiang Uygur Autonomous Region, China
| | - An-Jie Du
- Department of Ophthalmology, Yuncheng Central Hospital, Yuncheng 044000, Shanxi Province, China
| | - Yong Zhao
- Ophthalmic Center, No.474 Hospital of Chinese PLA, Urumqi 830013, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
143
|
Geevarghese A, Herman IM. Pericyte-endothelial crosstalk: implications and opportunities for advanced cellular therapies. Transl Res 2014; 163:296-306. [PMID: 24530608 PMCID: PMC3976718 DOI: 10.1016/j.trsl.2014.01.011] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/15/2014] [Accepted: 01/17/2014] [Indexed: 12/13/2022]
Abstract
Pericytes are mural cells of the microcirculation that have been shown to play key roles in regulating microvascular morphogenesis and stability throughout each tissue bed and organ system assessed. Of note, recent work has revealed that pericytes share several characteristics with mesenchymal- and adipose-derived stem cells, suggesting there may be lineage-related connections among bona fide pericytes and these vascular "progenitors," which can assume a perivascular position in association with endothelial cells. Hence, pericyte identity as a mediator of vascular remodeling may be confounded by its close relationships with its progenitors or pluripotent cell counterparts and yet demonstrates their potential utility as cell-based therapies for unmet clinical needs. Crucial to the development of such therapies is a comprehensive understanding of the origin and fate regulating these related cell types as well as the unveiling of the molecular mechanisms by which pericytes and endothelial cells communicate. Such mechanistic inputs, which disrupt normal cellular crosstalk during disease inception and progression, offer opportunities for intervention and are discussed in the context of the vasculopathies accompanying tumor growth, diabetes, and fibrosis.
Collapse
Affiliation(s)
- Anita Geevarghese
- Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Center for Innovations in Wound Healing Research, Tufts University School of Medicine, Boston, Mass
| | - Ira M Herman
- Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Center for Innovations in Wound Healing Research, Tufts University School of Medicine, Boston, Mass.
| |
Collapse
|
144
|
Davidson BA, Secord AA. Profile of pazopanib and its potential in the treatment of epithelial ovarian cancer. Int J Womens Health 2014; 6:289-300. [PMID: 24648773 PMCID: PMC3958497 DOI: 10.2147/ijwh.s49781] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological cancer. Recently, clinical trials have focused on novel antiangiogenic agents in combination with chemotherapy or alone in women with primary and recurrent ovarian cancer. Antiangiogenic agents include monoclonal antibodies, tyrosine-kinase inhibitors, and peptibodies. Many of these agents, including bevacizumab, pazopanib, nintedanib, cediranib, and trebananib, have been evaluated in randomized Phase III clinical trials, and all have demonstrated a progression-free survival (PFS) benefit. Specifically, maintenance pazopanib was shown to improve PFS in women with newly diagnosed EOC. Pazopanib, an oral TKI, inhibits several kinase receptors, including those for vascular endothelial growth factor (-1,-2,-3), platelet-derived growth factor (-α and -β), and fibroblast growth factor. It also targets stem cell-factor receptor (c-kit), interleukin 2-inducible T-cell kinase, lymphocyte-specific protein tyrosine kinase, and colony-stimulating factor 1 receptor. Pazopanib has been investigated in several Phase II and III clinical trials, with results indicating a potential role in the management of EOC. This article provides an overview of pazopanib in the treatment of EOC.
Collapse
Affiliation(s)
- Brittany A Davidson
- Division of Gynecologic Oncology, Duke Cancer Institute, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Angeles Alvarez Secord
- Division of Gynecologic Oncology, Duke Cancer Institute, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
145
|
Bachelot T, Garcia-Saenz JA, Verma S, Gutierrez M, Pivot X, Kozloff MF, Prady C, Huang X, Khosravan R, Wang Z, Cesari R, Tassell V, Kern KA, Blay JY, Lluch A. Sunitinib in combination with trastuzumab for the treatment of advanced breast cancer: activity and safety results from a phase II study. BMC Cancer 2014; 14:166. [PMID: 24606768 PMCID: PMC3995914 DOI: 10.1186/1471-2407-14-166] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 02/20/2014] [Indexed: 12/21/2022] Open
Abstract
Background This phase II study evaluated the efficacy and safety/tolerability of sunitinib plus trastuzumab in patients with HER2-positive advanced breast cancer (ABC). Methods Eligible patients received sunitinib 37.5 mg/day and trastuzumab administered either weekly (loading, 4 mg/kg; then weekly 2 mg/kg) or 3-weekly (loading, 8 mg/kg; then 3-weekly 6 mg/kg). Prior trastuzumab and/or lapatinib treatment were permitted. The primary endpoint was objective response rate (ORR). Results Sixty patients were enrolled and evaluable for safety; 57 were evaluable for efficacy. The majority of patients (58%) had received no prior chemotherapy in the metastatic setting. The ORR was 37%; the clinical benefit rate (CBR; percent objective response plus stable disease ≥ 24 weeks) was 56%. Among patients who were treatment-naïve or had received only adjuvant therapy, the ORR was 44% and the CBR was 59%. Overall, median overall survival had not been reached and the 1-year survival rate was 91%. The majority of adverse events (AEs) were mild to moderate in severity. Forty percent of patients experienced AEs related to measured left ventricular ejection fraction (LVEF) declines, which occurred more frequently in patients who had received prior anthracycline treatment. Ten percent of patients exhibited symptoms related to LVEF declines. One patient died on study from cardiogenic shock. Antitumor response and several safety parameters appeared to correlate with sunitinib exposure. Conclusions Sunitinib plus trastuzumab demonstrated antitumor activity in patients with HER2-positive ABC, particularly those who were treatment-naïve or had only received prior adjuvant treatment. Sunitinib plus trastuzumab had acceptable safety and tolerability in patients with HER2-positive ABC who had not received prior anthracycline therapy. Trial registration NCT00243503
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Jean-Yves Blay
- EORTC, Soft Tissue and Bone Sarcoma Group, Centre Léon-Bérard and Université Claude Bernard, Lyon, France.
| | | |
Collapse
|
146
|
Iqbal M, Ezzeldin E, Wani TA, Khalil NY. Simple, sensitive and rapid determination of linifanib (ABT-869), a novel tyrosine kinase inhibitor in rat plasma by UHPLC-MS/MS. Chem Cent J 2014; 8:13. [PMID: 24533632 PMCID: PMC3932137 DOI: 10.1186/1752-153x-8-13] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 02/12/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Linifanib (ABT-869) is an orally active receptor tyrosine kinase inhibitor, which simultaneously inhibits vascular endothelial and platelet derived growth factor receptor. The aim of the present study was to develop an UHPLC-MS/MS method for the quantification of linifanib in rat plasma to support the pharmacokinetic and toxicokinetic studies. RESULTS Linifanib was separated on Acquity UPLC BEH™ C18 column (50 × 2.1 mm, i.d. 1.7 μm) using acetonitrile-10 mM ammonium acetate (60:40, v/v) as an isocratic mobile phase at a flow rate of 0.3 mL/min with sunitinib as internal standard (IS). Detection was performed on tandem mass spectrometer using electrospray ionization source in positive mode by multiple reaction monitoring. The monitored transitions were set at m/z 376.05 > 250.97 for linifanib and m/z 399.12 >283.02 for IS, respectively. Both linifanib and IS were eluted at 0.68 and 0.44 min, respectively with a total run time of 2.0 min only. The calibration curve was found to be linear over the concentration range of 0.40-500 ng/mL. The intra- and inter-day precision value was ≤10.6% and the accuracy ranged from 90.9-108.9%. In addition, all the validation results were within general assay acceptability criteria according to guidelines of bio-analytical method validation. CONCLUSION A selective and sensitive UHPLC-MS/MS method was developed and validated for the determination of linifanib in rat plasma for the first time. The developed method is simple, sensitive and rapid in terms of chromatographic separation and sample preparation and was successfully applied in a pilot pharmacokinetic study in rats.
Collapse
Affiliation(s)
- Muzaffar Iqbal
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
- Bioavailability Laboratory, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, Saudi Arabia
| | - Essam Ezzeldin
- Bioavailability Laboratory, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, Saudi Arabia
| | - Tanveer A Wani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Nasr Y Khalil
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
147
|
Anti-angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis 2014; 17:471-94. [PMID: 24482243 PMCID: PMC4061466 DOI: 10.1007/s10456-014-9420-y] [Citation(s) in RCA: 518] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 01/15/2014] [Indexed: 12/17/2022]
Abstract
Tumours require a vascular supply to grow and can achieve this via the expression of pro-angiogenic growth factors, including members of the vascular endothelial growth factor (VEGF) family of ligands. Since one or more of the VEGF ligand family is overexpressed in most solid cancers, there was great optimism that inhibition of the VEGF pathway would represent an effective anti-angiogenic therapy for most tumour types. Encouragingly, VEGF pathway targeted drugs such as bevacizumab, sunitinib and aflibercept have shown activity in certain settings. However, inhibition of VEGF signalling is not effective in all cancers, prompting the need to further understand how the vasculature can be effectively targeted in tumours. Here we present a succinct review of the progress with VEGF-targeted therapy and the unresolved questions that exist in the field: including its use in different disease stages (metastatic, adjuvant, neoadjuvant), interactions with chemotherapy, duration and scheduling of therapy, potential predictive biomarkers and proposed mechanisms of resistance, including paradoxical effects such as enhanced tumour aggressiveness. In terms of future directions, we discuss the need to delineate further the complexities of tumour vascularisation if we are to develop more effective and personalised anti-angiogenic therapies.
Collapse
|
148
|
Kudelka MR, Grossniklaus HE, Mandell KJ. Emergence of dual VEGF and PDGF antagonists in the treatment of exudative age-related macular degeneration. EXPERT REVIEW OF OPHTHALMOLOGY 2014; 8:475-484. [PMID: 30613209 DOI: 10.1586/17469899.2013.840095] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Neovascular ('wet') age-related macular degeneration (AMD) is the leading cause of blindness among Caucasians over the age of 55 in the USA and is an important cause of ocular morbidity worldwide. Progress in oncology, and more recently ophthalmology, led to the development of VEGF antagonists, three of which are now approved for the treatment of wet AMD. Recent discoveries in ophthalmology and vascular biology, however, suggest that combined inhibition of VEGF and platelet-derived growth factor (PDGF) may be more beneficial than inhibition of VEGF alone. Accordingly, numerous studies are underway to evaluate the role of anti-VEGF/PDGF combination therapies for the treatment of wet AMD. This review discusses the biology of VEGF and PDGF and current preclinical and clinical data exploring the use of combined VEGF/PDGF inhibitors in the treatment of neovascular age-related macular degeneration.
Collapse
Affiliation(s)
- Matthew R Kudelka
- Department of Biochemistry, Emory University School of Medicine, 1510 Clifton Rd NE, Rollins Research Center - Room #4086, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
149
|
Lee SH, Colosi P, Lee H, Ohn YH, Kim SW, Kwak HW, Park TK. Laser photocoagulation enhances adeno-associated viral vector transduction of mouse retina. Hum Gene Ther Methods 2013; 25:83-91. [PMID: 24191872 DOI: 10.1089/hgtb.2013.089] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Laser photocoagulation is a well-established treatment modality for retinal disease. Discrete laser burns can be placed anywhere in the retina, singly or multiply, and the burn intensity is controllable. This study investigates the effect of prior laser photocoagulation on the retinal transduction properties of intravitreally administered adeno-associated viral (AAV) vectors. C57BL/6J mice were subjected to unilateral laser photocoagulation 48 hr before bilateral intravitreal injection of self-complementary cytomegaloviral enhanced green fluorescent protein (EGFP) vectors packaged in AAV type 2, 5, and 8 capsids. The eyes were enucleated 4 weeks after injection and examined by histochemistry and quantitative image analysis. Laser pretreatment resulted in substantially increased localized transduction around the burn site for all AAV capsid types. Without laser pretreatment, the vectors transduced only ganglion cells (AAV2) or sporadic cells around the optic nerve head (AAV5 and AAV8). Laser pretreatment increased AAV2 vector expression throughout the entire retina and focally at the burn site. Transduced cells at the burn site included retinal pigment epithelium (RPE), photoreceptors, Müller cells, inner nuclear layer cells, and retinal ganglion cells. The AAV5 vector showed increased RPE transduction at the burn site only. The AAV8 vector showed augmented expression in RPE, photoreceptors, and Müller cells around the burn site. Migrating RPE cells, present in the neural retina near the burn site, were also transduced by all three capsid types as evidenced by colocalization of EGFP and cytokeratin. Laser photocoagulation can be used to precisely direct AAV vector transduction to discrete locations in the retina. A combination of laser and AAV-mediated gene expression may allow the development of improved therapies for diabetic retinopathy, branch and central vein occlusion, and age-related macular degeneration.
Collapse
Affiliation(s)
- Si Hyung Lee
- 1 Department of Ophthalmology, Soonchunhyang University , College of Medicine, Bucheon 420-767, Korea
| | | | | | | | | | | | | |
Collapse
|
150
|
Chintalgattu V, Rees ML, Culver JC, Goel A, Jiffar T, Zhang J, Dunner K, Pati S, Bankson JA, Pasqualini R, Arap W, Bryan NS, Taegtmeyer H, Langley RR, Yao H, Kupferman ME, Entman ML, Dickinson ME, Khakoo AY. Coronary microvascular pericytes are the cellular target of sunitinib malate-induced cardiotoxicity. Sci Transl Med 2013; 5:187ra69. [PMID: 23720580 DOI: 10.1126/scitranslmed.3005066] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sunitinib malate is a multitargeted receptor tyrosine kinase inhibitor used in the treatment of human malignancies. A substantial number of sunitinib-treated patients develop cardiac dysfunction, but the mechanism of sunitinib-induced cardiotoxicity is poorly understood. We show that mice treated with sunitinib develop cardiac and coronary microvascular dysfunction and exhibit an impaired cardiac response to stress. The physiological changes caused by treatment with sunitinib are accompanied by a substantial depletion of coronary microvascular pericytes. Pericytes are a cell type that is dependent on intact platelet-derived growth factor receptor (PDGFR) signaling but whose role in the heart is poorly defined. Sunitinib-induced pericyte depletion and coronary microvascular dysfunction are recapitulated by CP-673451, a structurally distinct PDGFR inhibitor, confirming the role of PDGFR in pericyte survival. Thalidomide, an anticancer agent that is known to exert beneficial effects on pericyte survival and function, prevents sunitinib-induced pericyte cell death in vitro and prevents sunitinib-induced cardiotoxicity in vivo in a mouse model. Our findings suggest that pericytes are the primary cellular target of sunitinib-induced cardiotoxicity and reveal the pericyte as a cell type of concern in the regulation of coronary microvascular function. Furthermore, our data provide preliminary evidence that thalidomide may prevent cardiotoxicity in sunitinib-treated cancer patients.
Collapse
|