Oncology/endocrine
Inhibition of In Vivo Tumor Angiogenesis and Growth Via Systemic Delivery of an Angiopoietin 2-Specific RNA Aptamer

https://doi.org/10.1016/j.jss.2007.04.028Get rights and content

Background

Cellular events mediated by the Tie2 receptor are important to tumor neovascularization. Despite the complex interplay of the best-characterized Tie2 ligands, angiopoietins 1 and 2, Ang2 is purportedly “proangiogenic” in the presence of vascular endothelial growth factor. We examined whether in vivo administration of an RNA aptamer that specifically blocks Ang 2 would inhibit tumor angiogenesis and growth.

Methods

Ang2-mediated Tie2 receptor phosphorylation was assessed in vitro in the absence and presence of aptamer coupled to polyethylene glycol.

In vivo angiogenesis assay

CT26 murine colon carcinoma cells expressing green fluorescent protein were delivered into mouse dorsal skinfold window chambers. Animals received daily intraperitoneal injections of phosphate-buffered saline, low-dose (Ang2 aptamer-LD; 1 mg/kg/d), or high-dose aptamer (Ang2 aptamer-HD; 10 mg/kg/d). Vascular length density was measured under fluorescence microscopy.

Primary tumor growth

CT26 cells expressing luciferase were injected into flanks of BALB/c mice to allow tumor growth monitoring by bioluminescence imaging. Animals received continuous phosphate-buffered saline or aptamer (1 mg/kg/d) via ALZET pumps. Tumors were assessed for CD31/PECAM-1 immunostaining and Hoechst dye uptake.

Results

Pegylated aptamer inhibited Tie2 phosphorylation. Systemic aptamer administration reduced vascular length density (P ≤ 0.03) and decreased bioluminescence emission (P < 0.04), corresponding to 50% decrease in tumor volume (P = 0.04). Control tumors displayed abundant vascular marker staining, in contrast to tumors from aptamer-treated animals.

Conclusions

in vivo administration of a clinically relevant, pegylated RNA aptamer specifically designed against Ang2 inhibited tumor angiogenesis and growth. These findings support targeted Ang2 inhibition as a relevant anti-angiogenic, anti-neoplastic strategy.

Introduction

Angiogenesis is necessary for successful tumor growth. Although inhibition of vascular endothelial growth factor (VEGF) represents the most validated anti-angiogenic approach described thus far, VEGF works in concert with other components of the angiogenic process [1]. Of these, some of the most important are thought to be the tyrosine kinase receptor Tie2 (Tek), and its ligands, the angiopoietins [1]. Numerous investigations have underscored the significance of Tie2 and the angiopoietins in normal and pathological angiogenesis [1]. The Tie2/angiopoietin (Ang) system comprises a critical signal transduction pathway such that mice deficient in Tie2 exhibit impaired vascular remodeling, disrupted endothelial-mesenchymal interactions, and resultant embryonic lethality [1, 2, 3, 4]. Tie2 is present predominantly, but not exclusively, on endothelial cells during both developmental and adult stages, and its expression is increased during adult vascularization [1, 2]. Ang1 is constitutively expressed by pericytes and smooth muscle cells, and is thought to primarily promote interactions between endothelial and support cells to stabilize the developing vasculature [5, 6, 7]. Effects of Ang1 are antagonized by Ang2, which is expressed at sites of vascular remodeling in adults [8, 9]. Expression of Ang1 in quiescent vasculature and that of Ang2 at sites of active angiogenesis had given rise to the hypothesis that angiopoietins 1 and 2 have opposite functions in vascular development. More recent studies have challenged this concept and emphasized the context in which angiopoietins operate [10, 11, 12, 13, 14]. For example, Ang1 has been shown to act as a negative regulator of VEGF-induced angiogenesis in the heart [13]. Ang2, on the other hand, has been reported to enhance angiogenesis in the presence of VEGF, but to promote vessel regression in its absence [10, 11, 12, 13].

Along with a growing body of knowledge on vascular development, the role of Tie2 and the angiopoietins in angiogenesis and tumor development is becoming better characterized [15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25]. Animal studies have shown Tie2 receptor blockade to inhibit angiogenesis, tumor growth, and metastasis [18, 19, 20, 21, 22]. In a murine model of ischemia-induced retinopathy, systemic administration of an adenovirus encoding the extracellular domain of Tie2 (AdExTek) inhibited retinal neovascularization [17]. Lin et al. demonstrated that administering AdExTek to mice significantly inhibited subcutaneous tumor growth, and intravenous co-injection of tumor cells and AdExTek reduced pulmonary metastatic potential [18]. Human melanoma cells expressing the extracellular domain of Tie2 were shown to inhibit angiogenesis and tumor growth in mouse xenografts [19]. Work specifically directed at the angiopoietins including overexpression studies showed Ang1 to inhibit tumor angiogenesis or growth, and Ang2 to enhance it [22, 23, 25]. Human colon cancer cells overexpressing Ang1 implanted into nude mouse livers inhibited tumor angiogenesis and produced significantly smaller tumors, suggesting potential relevance of the Tie2/angiopoietin pathway in treating metastatic colorectal cancer [25]. Consistent with the notion that the interplay among the angiopoietins and Tie2 dictates vascular stability, Zhang et al. demonstrated VEGF-mediated up-regulation of Ang2 with resultant pericyte loss and instability of host vessels adjacent to tumors, supporting a pro-angiogenic role [23].

Recently, angiogenesis in a non-tumor-bearing rat corneal pocket angiogenesis assay was inhibited by the local delivery of an RNA aptamer specifically designed to bind and inhibit Ang2 [24]. Based on this finding, an Ang2-specific aptamer was similarly synthesized based on the sequence and modifications described by White et al. [24], for use in our study.

By directly inhibiting protein activity, aptamers act similarly to monoclonal antibodies or small molecule drugs, increasing their potential clinical applicability [25, 26, 27, 28, 29, 30, 31]. Advantages of aptamers over monoclonal antibodies include the ability for chemical synthesis in large quantities and low immunogenicity in vivo, while exhibiting high affinity for target proteins [27, 32]. In addition to a therapeutic role, aptamers can be used to escort therapeutic or diagnostic reagents [33]. Aptamers targeting any protein or small molecule can be produced using in vitro selection processes. The Ang2-specific aptamer used in this study had been positively selected for recombinant human Ang2, and negatively selected against recombinant human Ang1 [24]. Optimization of a desired sequence for in vivo use requires manipulating the factors that dictate bioavailability, such as stability in biological fluids and systemic clearance [34]. The former, reliant on nucleic acid backbone, is enhanced by replacing ribonucleotides with 2′-amino, -fluoro, or -O-alkyl nucleotides [27, 35, 36]. Such modifications extend in vitro plasma half-lives of RNA oligonucleotides from seconds to 5 to 15 h. Exonuclease degradation in plasma can be prevented by capping the 3′ end of an oligonucleotide and further reinforced by replacing ribose and deoxyribose with modified nucleotides or non-nucleotide linkers [34]. To allow for practical chemical synthesis, aptamers are currently truncated to fewer than 40 nucleotides [34], promoting rapid systemic clearance. This can be overcome by attaching the aptamer to a polyethylene glycol (PEG) moiety or a liposomal surface [26, 37]. The clinical relevance of aptamers is brought to light by the use of VEGF aptamers in clinical trials for treatment of macular degeneration [38, 39].

It is not known whether inhibiting Tie2 or targeting the angiopoietins is more apt to generate a potent antineoplastic response. We hypothesized that systemic administration of a nuclease-resistant Ang2-specific aptamer, conjugated to 40 kDa PEG to enhance bioavailability, would impair angiogenesis and inhibit growth of primary tumors.

Section snippets

Ang2-Specific Aptamer

The sequence (GAGGACGAUGCGGACUAGCCUCAUCAGCUCAUGUGCCCCUC) had been previously shown by White et al. to bind and inhibit Ang2 function with high affinity and specificity, without affecting Ang1-mediated Tie2 activation [24]. To improve bioavailability, the aptamer was constructed using 41 2′-fluoro pyrimidines and 2′-hydroxy-purines, flanked by 5′ 40 kDa PEG and C6 aminolinker, and 3′ inverted deoxythymidine (Transgenomic, Inc., Omaha, NE). The lyophilized oligonucleotide was suspended in

Effect of Pegylated Aptamer on Ang2-Mediated Tie2 Activity

Ang2 is known to phosphorylate Tie2 that is ectopically expressed in non-endothelial cell lines [9]. The RNA aptamer had previously been shown to inhibit Tie2 phosphorylation by Ang2 in vitro [24]. To verify that following conjugation to 40 kDa PEG the aptamer would inhibit Ang2, immunoprecipitation and WB were performed to detect Tie2 receptor phosphorylation. Two Hundred ninety-three cells stably expressing human Tie2 were treated with Ang2 in the absence and presence of molar excesses of the

Discussion

In an effort to determine if specific targeting of angiopoietin-2 in vivo would affect angiogenesis or tumor growth, we systemically administered a clinically-relevant, nuclease-resistant Ang2-specific RNA aptamer to mice bearing either dorsal window chambers or subcutaneous colorectal tumors. The aptamer had been previously shown to bind Ang2 with high affinity in vitro, and to inhibit angiogenesis in a rat corneal pocket assay when locally delivered [24]. To extend its in vivo applicability,

References (43)

  • G.D. Yancopoulos et al.

    Vascular-specific growth factors and blood vessel formation

    Nature

    (2000)
  • D.J. Dumont et al.

    Tek, a novel tyrosine kinase gene located on mouse chromosome 4, is expressed in endothelial cells and their presumptive precursors

    Oncogene

    (1992)
  • T.N. Sato et al.

    Distinct roles of the receptor tyrosine kinases Tie-1 and Tie-2 in blood vessel formation

    Nature

    (1995)
  • D.J. Dumont et al.

    Dominant-negative and targeted null mutations in the endothelial receptor tyrosine kinase, tek, reveal a critical role in vasculogenesis of the embryo

    Genes Dev

    (1994)
  • A. Uemura et al.

    Recombinant angiopoietin-1 restores higher-order architecture of growing blood vessels in mice in the absence of mural cells

    J Clin Invest

    (2002)
  • G. Thurston et al.

    Leakage-resistant blood vessels in mice transgenically overexpressing angiopoietin-1

    Science

    (1999)
  • P. Maisonpierre et al.

    Angiopoietin-2, a natural antagonist for Tie2 that disrupts n vivo angiogenesis

    Science

    (1997)
  • J. Holash et al.

    Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF

    Science

    (1999)
  • I.B. Lobov et al.

    Angiopoietin-2 displays the VEGF-dependent modulation of capillary structure and endothelial survival in vivo

    Proc Natl Acad Sci

    (2002)
  • T. Korff et al.

    Blood vessel maturation in a 3-dimensional spheroidal co-culture model: Direct contact with smooth muscle cells regulates endothelial cell quiescence and abrogates VEGF responsiveness

    FASEB J

    (2001)
  • R.P. Visconti et al.

    Orchestration of angiogenesis and arteriovenous contribution by angiopoietins and vascular endothelial growth factor (VEGF)

    Proc Natl Acad Sci

    (2002)
  • Cited by (56)

    • Dynamic angiopoietin-2 assessment predicts survival and chronic course in hospitalized patients with COVID-19

      2021, Blood Advances
      Citation Excerpt :

      All the evidence suggests that angiopoietin-2 blockade is an important therapeutic option, as the drugs currently used for COVID-19 achieve low survival benefit. Although this treatment approach has not been examined in a human study, many methods, including RNA aptamers,21,22 blocking antibodies and peptides,23 and angiopoieitin-1 administration24 have been used to block angiopoietin-2 in animal models. More recently, Lomas-Neira et al25 used the small interfering RNA inhibition of angiopoietin-2 protein synthesis to block its activity in an experimental mouse model of ARDS.

    • In Vivo Selection Against Human Colorectal Cancer Xenografts Identifies an Aptamer That Targets RNA Helicase Protein DHX9

      2016, Molecular Therapy Nucleic Acids
      Citation Excerpt :

      Specificity for cancer cells is necessary to minimize toxicity to normal tissues. Aptamers are a class of artificial oligonucleotides with the ability to fold into precise three-dimensional and globular confirmations that enable them to directly and specifically bind to targeted proteins, and are thus ideal for targeted molecular therapy.2,3,4 The Systematic Evolution of Ligands by Exponential Enrichment (SELEX) method is typically applied to select aptamers in vitro against purified protein, complex-protein mixtures or whole cells.5,6,7

    • The angiopoietin: TIe 2 interaction: A potential target for future therapies in human vascular disease

      2013, Cytokine and Growth Factor Reviews
      Citation Excerpt :

      In preclinical models, blocking the activity of ang2 specifically has demonstrated efficacy in the inhibition of the growth and angiogenesis of tumours. Methods used include the use of ang2 blocking monoclonal antibodies [143,144], high-affinity nuclease resistant RNA ligands (aptamers) [145], peptide-Fc fusion proteins which increase the stability of ang2-binding peptides (peptibodies) [146–150], and a bivalent ang2-binding peptide-antibody fusion protein, known as a CoxV-Body [151]. The fusion of two ang2-binding peptides to a monoclonal antibody results in a preservation of its ability to bind ang2, but with a greatly extended in vivo half-life.

    • VEGF and angiopoietin signaling in tumor angiogenesis and metastasis

      2011, Trends in Molecular Medicine
      Citation Excerpt :

      A soluble Tie2 extracellular domain-containing fusion protein, which binds both Ang1 and Ang2, was initially shown to diminish tumor growth and lung metastasis in syngeneic mouse tumor models [140]. More recently, selective peptibody [141] or antibody [142] inhibitors of Ang2, as well as an Ang2-specific aptamer [143] (Table 1), were shown to reduce tumor growth in numerous mouse models, suggesting that Ang2 is the main angiopoietin ligand in tumors. In addition, an Ang2 CovX-Body, composed of a peptide coupled to a monoclonal antibody via covalent linker, inhibited tumor vascularization and growth by preventing Ang2 binding to Tie2 [144], and is currently being tested in phase II clinical trials (www.clinicaltrials.gov).

    View all citing articles on Scopus
    View full text