Vascular endothelial growth factor (VEGF) blockers have been developed for the treatment of proliferative diabetic retinopathy (PDR), the leading cause of visual impairments in the working-age population in the Western world. However, limitations to anti-VEGF therapies may exist because of the local production of other proangiogenic factors that may cause resistance to anti-VEGF interventions. Thus, novel therapeutic approaches targeting additional pathways are required. Here, we identified a sulfated derivative of the Escherichia coli polysaccharide K5 [K5-N,OS(H)] as a multitarget molecule highly effective in inhibiting VEGF-driven angiogenic responses in different in vitro, ex vivo, and in vivo assays, including a murine model of oxygen-induced retinopathy. Furthermore, K5-N,OS(H) binds a variety of heparin-binding angiogenic factors upregulated in PDR vitreous humor besides VEGF, thus inhibiting their biological activity. Finally, K5-N,OS(H) hampers the angiogenic activity exerted in vitro and in vivo by human vitreous fluid samples collected from patients with PDR. Together, the data provide compelling experimental evidence that K5-N,OS(H) represents an antiangiogenic multitarget molecule with potential implications for the therapy of pathologic neovessel formation in the retina of patients with PDR.

Proliferative diabetic retinopathy (PDR) is the main cause of blindness in the working-age population (1). Numerous angiogenic factors and cytokines related to retinal neovascularization have been identified (25) whose levels are elevated in the ocular fluid of patients with PDR (57). Among them, upregulation of vascular endothelial growth factor (VEGF) has been associated with the breakdown of the blood-retina barrier, increased vessel permeability, and retinal neovascularization (810).

Three different VEGF blockers have evolved over time for PDR therapy (pegaptanib, ranibizumab, and aflibercept); a fourth agent, bevacizumab, is used off label (11). Even though anti-VEGF interventions have shown better outcomes than alternative treatments, limitations to anti-VEGF therapies may exist, including short duration of action, local and systemic adverse effects, and poor response in a significant percentage of patients (1214). Furthermore, the production of other angiogenic factors may cause resistance to anti-VEGF therapies. Thus, the identification of novel therapeutic approaches targeting additional pathways is warranted (15,16).

Heparan sulfates (HS) are N,O-sulfated glycosaminoglycans (GAGs) present on the cell surface and extracellular matrix as HS proteoglycans (HSPGs). HSPGs modulate the activity of heparin-binding angiogenic growth factors, including VEGF and fibroblast growth factor (FGF) family members, by mediating their interaction with signaling receptors (17,18). Thus, synthetic or chemically modified heparin/HS-like molecules may act as angiogenic factor inhibitors, hampering their binding to endothelial receptors (19). Relevant to PDR, intravitreal injection of high concentrations of heparin/HS or heparin analogs reduces retinal neovascularization in a model of oxygen-induced retinopathy (OIR) (20,21), originally developed in mice as a model of retinopathy of prematurity (22) and widely adopted also to study angiogenesis in PDR (23).

The Escherichia coli K5 polysaccharide, whose name coincides with the abbreviation of the structurally and functionally unrelated antiangiogenic plasminogen proteolytic fragment kringle 5 (24), has the same structure as the heparin precursor N-acetyl heparosan (25). K5 polysaccharide derivatives, produced by chemical and enzymatic modifications, represent a new class of heparin/HS-like compounds (26). In particular, the highly N,O-sulfated derivative K5-N,OS(H) is endowed with a negligible anticoagulant activity and acts as an antiangiogenic, anti-inflammatory, and antiviral molecule due to its capacity to interact with different heparin-binding proteins (26,27). Thus, K5 derivatives may represent a novel class of multitarget molecules able to interact with and block the activity of a broad range of heparin-binding angiogenic factors involved in the pathogenesis of PDR.

Here, we investigated the capacity of K5-N,OS(H) to affect the angiogenic activity of the major heparin-binding VEGF-A165 isoform (hereafter named VEGF) expressed in PDR (3). The results demonstrate that K5-N,OS(H) inhibits VEGF-driven neovascular responses in vitro, ex vivo, and in vivo. Furthermore, K5-N,OS(H) binds a variety of heparin-binding angiogenic factors present in the vitreous humor of patients with PDR and inhibits the angiogenic activity of PDR vitreous fluid samples. Thus, K5-N,OS(H) represents an antiangiogenic multitarget molecule with potential therapeutic implications in PDR.

Reagents

K5 derivatives (28) and unfractionated heparin were from Glycores 2000; M199 medium and FCS from Gibco Life Technologies (Grand Island, NY); FGF2 from Tecnogen (Caserta, Italy); connective tissue growth factor (CTGF) and stromal cell–derived factor-α (SDF1α) from PeproTech (Rock Hill, NJ); hepatocyte growth factor (HGF), IGF-1, interleukin-8 (IL8), and platelet-derived growth factor-B (PDGF-B) from ReliaTech (Wolfenbüttel, Germany); high mobility group box-1 (HMGB1) from ProSpec (Rehovot, Israel); bevacizumab from Roche (Basel, Switzerland); and cyclopeptide vascular endothelial growth inhibitor (cyclo-VEGI) and the soluble form of VEGFR2 (sVEGFR2) from Calbiochem (Darmstadt, Germany). VEGF (VEGF-A165 isoform) was provided by K. Ballmer-Hofer (PSI, Villigen, Switzerland). Anti-VEGFR2, anti-pVEGFR2, and anti-rabbit horseradish peroxidase (HRP)-labeled antibodies were from Santa Cruz Biotechnology (Santa Cruz, CA); rat monoclonal anti-CD31 antibody from BD Biosciences (San Diego, CA); Alexa Fluor 488 from Molecular Probes (Eugene, OR); and anti–β-actin antibody from Sigma-Aldrich (St. Louis, MO).

Surface Plasmon Resonance Analysis

Biotinylated heparin was immobilized onto a Biacore SA sensor chip (GE Healthcare, Madison, WI) containing pre-immobilized streptavidin, whereas sVEGFR2 was immobilized on an activated CM5 sensor chip (29). For competition experiments, VEGF (150 nmol/L) plus increasing GAG concentrations were injected over heparin or sVEGFR2 surfaces, and bound analyte was measured (resonance units [RU]) at the end of injection. For evaluation of K5-N,OS(H)/growth factor interaction, proteins were injected over the heparin surface in the presence of increasing GAG concentrations.

Animals

Procedures involving animals were performed in agreement with the ARVO Statement for the Use of Animals in Ophthalmic and Vision Research and in compliance with Italian guidelines for animal care (DL 116/92) and European Communities Council Directive (86/609/EEC). Procedures were approved by the Ethical Committee in Animal Experiments of the universities involved in the study.

VEGF-Mediated Cell-Cell Adhesion Assay

GAG-deficient A745 CHO cells were transfected with pcDNA3 harboring the human VEGFR2 extracellular domain (ECD) conjugated with yellow fluorescent protein (YFP) to generate stable ECD-VEGFR2-YFP transfectants. CHO-K1 cells were seeded in 24-well plates at 50,000 cells/cm2. After 24 h, ECD-VEGFR2-YFP cells (50,000 cells/cm2) were added to CHO-K1 monolayers in the presence of 75 ng/mL VEGF and different competitors. After 2 h at 4°C, ECD-VEGFR2-YFP cells bound to the CHO-K1 monolayer were counted.

VEGFR2 Phosphorylation and Proliferation Assays

Human umbilical vein endothelial cells (HUVECs) treated with 5.0 ng/mL VEGF and increasing concentrations of K5-N,OS(H) were probed by Western blotting with anti-pVEGFR2 antibody. For the cell proliferation assay, HUVECs (15,000 cells/cm2) were treated with 10 ng/mL VEGF or vitreous fluid diluted in culture medium (1:4, volume:volume [vol:vol]) plus 2.5% FCS and increasing K5-N,OS(H) concentrations. After 24 h, cells were counted.

HUVEC Sprouting Assay

HUVEC aggregates were embedded in fibrin gel (30) and treated with 30 ng/mL VEGF or diluted vitreous fluid (1:4, vol:vol) plus increasing K5-N,OS(H) concentrations. Sprouts were counted after 24 h.

Wounding of HUVEC Monolayer

Cell monolayers made quiescent by overnight incubation in medium with 3.5% FCS were wounded with a 1.0-mm-wide rubber policeman. Then, cells were incubated in fresh medium with 3.5% FCS and 50 ng/mL of the angiogenic factor under test in the absence or presence of 100 ng/mL K5-N,OS(H). After 24 h, wound repair was quantified by computerized analysis.

Murine Aorta Ring Assay

Fibrin-embedded aortic rings were incubated in serum-free endothelial cell basal medium (Clonetics) with 30 ng/mL VEGF and increasing K5-N,OS(H) concentrations. After 5 days, vessel sprouts were counted under a stereo microscope (31).

Ex Vivo Murine Retina Angiogenesis Assay

Fibrin-embedded retina fragments (32,33) were treated with 75 ng/mL VEGF and increasing K5-N,OS(H) concentrations. At day 7, vessel sprouts were counted.

Chick Embryo Chorioallantoic Membrane Assay

Alginate beads (3 µL) containing 2 µL of vitreous fluid or 100 ng/pellet of VEGF and increasing amounts of K5-N,OS(H) were placed on the chorioallantoic membrane (CAM) of fertilized white Leghorn chicken eggs at day 11 (30). After 3 days, microvessels converging toward the implant were counted.

Rabbit Cornea Assay

Polymer Elvax-40 pellets containing 200 ng VEGF with or without 100 ng K5-N,OS(H) were implanted in the cornea of male New Zealand white rabbits (n = 9; Charles River) (34). Angiogenic score (vessel density × distance in millimeters) was calculated at day 11.

Murine OIR Model

Experiments were performed on C57BL/6J mouse pups (22). Sixteen pups were used for experimental protocol setup, and five pups per group were randomly selected for K5-N,OS(H) treatment followed by Western blotting, immunohistochemistry, or quantitative RT-PCR analysis (71 pups total). Litters of mouse pups with their nursing mothers were room-air reared or exposed to 75 ± 2% oxygen between postnatal day (PD)7 and PD12 before return to room air. At PD12 and PD15, 1.0 µL K5-N,OS(H) in sterile PBS (from 0.06 to 60 ng/µL) was intravitreally injected into the left eye, whereas the right eye was injected with vehicle. At PD17, retinal extracts were probed by Western blotting with anti-pVEGFR2, anti-Bax, anti–Bcl-2, and anti–cytochrome c antibodies, normalizing their levels to those of VEGFR2 or β-actin (35). Also, quantitative RT-PCR analysis was performed on total RNA extracted from PD17 retinas, and data were analyzed by the formula 2-ΔΔCT using Rpl13a gene expression as internal standard (35). Oligonucleotide primers were as follows: Vegf-A forward: 5′-GCACATAGGAGAGATGAGCTTCC-3′ and reverse: 5′-CTCCGCTCTGAACAAGGCT-3′; Igf-1 forward: 5′-TTCAGTTCGTGTGTGGACCGAG-3′ and reverse: 5′-TCCACAATGCCTGTCTGAGGTG-3′; Fgf-2 forward: 5′-GCGACCCACACGTCAAACTA-3′ and reverse: 5′-TCCCTTGATAGACACAACTCCTC-3′; angiopoietin-2 forward: 5′-GACTTCCAGAGGACGTGGAAAG-3′ and reverse: 5′-CTCATTGCCCAGCCAGTACTC-3′; erythropoietin forward: 5′-ACTCTCCTTGCTACTGATTCCT-3′ and reverse: 5′-ATCGTGACATTTTCTGCCTCC-3′; Tnfα forward: 5′-GCCTCTTCTCATTCCTGCTT-3′ and reverse: 5′-CTCCTCCACTTGGTGGTTTG-3′; Il-1β forward: 5′-TCCTTGTGCAAGTGTCTGAAGC-3′ and reverse: 5′-ATGAGTGATACTGCCTGCCTGA-3′; Il-6 forward: 5′-TCTGCAAGAGACTTCCATCCAGT-3′ and reverse: 5′-TCTGCAAGTGCATCATCGTTGT-3′; iNos forward: 5′-GGCAAACCCAAGGTCTACGTT-3′ and reverse: 5′-TCGCTCAAGTTCAGCTTGGT-3′; Rpl13a forward: 5′-CACTCTGGAGGAGAAACGGAAGG-3′ and reverse: 5′-GCAGGCATGAGGCAAACAGTC-3′.

Immunohistochemical analysis of CD31+ retinal vasculature and quantification of preretinal neovascular tufts were performed on retinal whole mounts (36).

Human Vitreous Fluid Samples

Patients with PDR (Table 1) underwent pars plana vitrectomy at the Clinics of Ophthalmology of the University of Brescia. Collection and analysis of human samples was approved by the internal review board of the Spedali Civili of Brescia and followed the principles of the Declaration of Helsinki. In some patients, intravitreal injection of the VEGF blockers bevacizumab or ranibizumab was performed 10–15 days prior to vitrectomy (Table 1). Samples were stored at −80°C.

Table 1

Patients with PDR

Patients for pooled vitreous humor assaysPatients for individual vitreous humor assays
Patients/eyes 19/20 10/10 
Sex (male) 13/19 7/10 
Age (years, mean ± SD) 64 ± 12 66 ± 10 
Type 1/type 2 diabetes 4/15 0/10 
Insulin treatment 11 
Nephropathy 
Hypertension 17 10 
Dyslipidemia 
Ophthalmology features 
 PDR 20 10 
 PDR with vitreous hemorrhage 19 
 PDR with macular edema 14 
Therapies 
 Intravitreal injection of anti-VEGF blocker 12 
 Panretinal laser photocoagulation 17 
Patients for pooled vitreous humor assaysPatients for individual vitreous humor assays
Patients/eyes 19/20 10/10 
Sex (male) 13/19 7/10 
Age (years, mean ± SD) 64 ± 12 66 ± 10 
Type 1/type 2 diabetes 4/15 0/10 
Insulin treatment 11 
Nephropathy 
Hypertension 17 10 
Dyslipidemia 
Ophthalmology features 
 PDR 20 10 
 PDR with vitreous hemorrhage 19 
 PDR with macular edema 14 
Therapies 
 Intravitreal injection of anti-VEGF blocker 12 
 Panretinal laser photocoagulation 17 

Data are n unless otherwise indicated.

K5 Derivatives Inhibit HSPG/VEGF/VEGFR2 Ternary Complex Formation

HSPGs mediate the engagement of VEGF with its signaling receptor VEGFR2, leading to the formation of HSPG/VEGF/VEGFR2 ternary complexes (18). To assess the impact of VEGF blockers on the formation of this bioactive complex, we developed a novel assay in which VEGF mediates cell-cell adhesion events due to its simultaneous interaction with VEGFR2 and HSPGs expressed in trans on neighboring cells (28). As shown in Fig. 1A, VEGF allows the dose-dependent adhesion of HSPG-deficient CHO cells overexpressing the ECD of VEGFR2 (ECD-VEGFR2-YFP cells) when seeded on a monolayer of HSPG-bearing CHO-K1 cells. Specificity of the interaction was demonstrated by the lack of activity of the unrelated heparin-binding FGF2 or of the VEGF-A121 isoform devoid of the heparin-binding domain. Furthermore, HSPG/VEGF/VEGFR2 complex formation was prevented by the anti-VEGF antibody bevacizumab and by the VEGFR2 antagonist cyclo-VEGI peptide (37) (Fig. 1B). Thus, cell-cell adhesion in this model is VEGF dependent and due to the formation in trans of HSPG/VEGF/VEGFR2 ternary complexes on neighboring cells.

Figure 1

Effect of K5 derivatives on HSPG/VEGF/VEGFR2 complex formation. A: HSPG-deficient ECD-VEGFR2-YFP A745 CHO transfectants were added to CHO-K1 monolayers in the presence of increasing concentrations of VEGF (black squares), 150 ng/mL VEGF-A121 (white squares), or 150 ng/mL FGF2 (black circles). B: ECD-VEGFR2-YFP cells were added to CHO-K1 monolayers in the presence of 75 ng/mL VEGF and increasing concentrations of bevacizumab (left panel) or cyclo-VEGI (right panel). C: ECD-VEGFR2-YFP cells were added to CHO-K1 monolayers in the presence of 75 ng/mL VEGF and increasing concentrations of K5 (black squares), K5-NS (white squares), K5-N,OS(L) (black circles), K5-N,OS(H) (white circles), or heparin (black triangles). In all the experiments, adherent cells were photographed and counted after 2 h of incubation at 4°C. D: Representative photographs of fluorescent ECD-VEGFR2-YFP cells adherent on a CHO-K1 monolayer in the absence (a) or in the presence (b) of 1.0 µg/mL K5-N,OS(H). E and F: SPR analysis. VEGF was coinjected with increasing concentrations of K5 (black squares), K5-NS (white squares), K5-N,OS(L) (black circles), K5-N,OS(H) (white circles), or heparin (black triangles) on Biacore sensor chips coated with immobilized heparin (E) or sVEGFR2 (F). The response (in RU) was recorded at the end of injection and plotted as a function of GAG concentration. Data are the mean ± SEM of three independent experiments.

Figure 1

Effect of K5 derivatives on HSPG/VEGF/VEGFR2 complex formation. A: HSPG-deficient ECD-VEGFR2-YFP A745 CHO transfectants were added to CHO-K1 monolayers in the presence of increasing concentrations of VEGF (black squares), 150 ng/mL VEGF-A121 (white squares), or 150 ng/mL FGF2 (black circles). B: ECD-VEGFR2-YFP cells were added to CHO-K1 monolayers in the presence of 75 ng/mL VEGF and increasing concentrations of bevacizumab (left panel) or cyclo-VEGI (right panel). C: ECD-VEGFR2-YFP cells were added to CHO-K1 monolayers in the presence of 75 ng/mL VEGF and increasing concentrations of K5 (black squares), K5-NS (white squares), K5-N,OS(L) (black circles), K5-N,OS(H) (white circles), or heparin (black triangles). In all the experiments, adherent cells were photographed and counted after 2 h of incubation at 4°C. D: Representative photographs of fluorescent ECD-VEGFR2-YFP cells adherent on a CHO-K1 monolayer in the absence (a) or in the presence (b) of 1.0 µg/mL K5-N,OS(H). E and F: SPR analysis. VEGF was coinjected with increasing concentrations of K5 (black squares), K5-NS (white squares), K5-N,OS(L) (black circles), K5-N,OS(H) (white circles), or heparin (black triangles) on Biacore sensor chips coated with immobilized heparin (E) or sVEGFR2 (F). The response (in RU) was recorded at the end of injection and plotted as a function of GAG concentration. Data are the mean ± SEM of three independent experiments.

Close modal

On this basis, different K5 derivatives chemically sulfated in the N position or N,O positions (Table 2) were investigated for the capacity to affect HSPG/VEGF/VEGFR2 complex formation in this assay. Similar to unmodified heparin, the highly N,O-sulfated K5-N,OS(H) prevents the adhesion of ECD-VEGFR2-YFP transfectants to HSPG-bearing cells; the low N,O-sulfated derivative K5-N,OS(L) was ∼20 times less potent than K5-N,OS(H), whereas K5-NS and K5 were ineffective (Fig. 1C and D).

Table 2

Chemical features and VEGF binding activity of K5 derivatives

GAGSO3/COO ratioAverage molecular weight (Da)VEGF-mediated cell-cell adhesion ID50 (µg/mL)SPR
VEGF/heparin binding ID50 (µg/mL)VEGF/sVEGFR2 binding ID50 (µg/mL)
K5 30,000 No inhibition No inhibition No inhibition 
K5-NS 1.00 15,000 >100 4.5 12.0 
K5-N,OS(L) 1.70 13,000 10.2 5.0 9.5 
K5-N,OS(H) 3.77 15,000 0.5 0.005 0.2 
Heparin 2.14 13,600 0.2 0.4 1.0 
GAGSO3/COO ratioAverage molecular weight (Da)VEGF-mediated cell-cell adhesion ID50 (µg/mL)SPR
VEGF/heparin binding ID50 (µg/mL)VEGF/sVEGFR2 binding ID50 (µg/mL)
K5 30,000 No inhibition No inhibition No inhibition 
K5-NS 1.00 15,000 >100 4.5 12.0 
K5-N,OS(L) 1.70 13,000 10.2 5.0 9.5 
K5-N,OS(H) 3.77 15,000 0.5 0.005 0.2 
Heparin 2.14 13,600 0.2 0.4 1.0 

Sulfate/carboxyl ratio (SO3/COO) and molecular weight values are from Leali et al. (28). GAGs were assessed for the capacity to inhibit the formation of HSPG/VEGF/VEGFR2 ternary complexes in a VEGF-mediated cell-cell adhesion assay and to prevent the binding of VEGF to heparin and sVEGFR2 immobilized to Biacore sensor chips.

To assess whether this activity was due to the ability of K5 derivatives to affect HSPG/VEGF and/or VEGF/VEGFR2 interactions, GAGs were investigated by surface plasmon resonance (SPR) analysis for their capacity to prevent the binding of VEGF to heparin or sVEGFR2 immobilized to Biacore sensor chips, a VEGF interaction that occurs with Kd values equal to 100 (38) and 3.0 nmol/L (39) for the two molecules, respectively. As shown in Fig. 1E and F, K5-N,OS(H) inhibits the binding of VEGF to immobilized heparin and sVEGFR2, being more effective than free heparin. At variance, K5-N,OS(L) and K5-NS exerted an inhibitory effect only at high doses and K5 was ineffective. The ID50 values calculated for K5 derivatives in cell-cell adhesion and SPR assays are summarized in Table 2.

In conclusion, K5-N,OS(H) acts as a potent VEGF antagonist able to prevent its interaction with heparin/HSPGs and VEGFR2, hampering the formation of bioactive HSPG/VEGF/VEGFR2 complexes. On this basis, the capacity of K5-N,OS(H) to inhibit the biological activity of VEGF was assessed in a series of angiogenesis bioassays.

K5-N,OS(H) Inhibits the Angiogenic Activity of VEGF

The antiangiogenic potential of K5-N,OS(H) was assessed in vitro, ex vivo, and in vivo on the endothelium of different species and tissue origin when stimulated by an optimal dose of VEGF able to elicit the maximal response in each assay.

K5-N,OS(H) inhibits VEGF-induced VEGFR2 phosphorylation in HUVECs, maximum effect being observed at 50 ng/mL of the GAG (Fig. 2A). Accordingly, a significant decrease in the cell proliferation rate was observed when HUVECs were treated with VEGF in the presence of K5-N,OS(H) (Fig. 2B). Also, K5-N,OS(H) prevents VEGF-mediated endothelial cell sprouting of fibrin-embedded HUVEC spheroids, murine aortic rings, and murine retinal fragments (Fig. 2C–E).

Figure 2

K5-N,OS(H) inhibits the angiogenic activity of VEGF. A: HUVECs were incubated for 10 min at room temperature in the absence (control) or presence of 5.0 ng/mL VEGF and increasing concentrations of K5-N,OS(H), lysed, and probed in a Western blot with anti-pVEGFR2 (Y1175) antibody. Uniform loading of the gel was confirmed using an antifocal adhesion kinase (FAK) antibody. B: HUVECs were grown with 10 ng/mL VEGF in the absence (black bars) or presence (white bars) of 10 ng/mL K5-N,OS(H) and counted after 24 h. Cell proliferation was expressed as fold increase in respect to the control. Data are the mean ± SEM of three independent determinations. C: HUVEC spheroids were embedded in fibrin gel and incubated with 30 ng/mL VEGF in the absence (black bars) or presence (white bars) of 10 ng/mL K5-N,OS(H). Formation of radially growing cell sprouts was evaluated after 24 h of incubation. Data are the mean ± SEM of three independent determinations. D: Murine aortic rings were embedded in fibrin gel and incubated in the presence of VEGF (30 ng/mL) and increasing concentrations of K5-N,OS(H). After 5 days, vessel sprouts were counted under a stereo microscope. Data are the mean ± SEM of three independent determinations. E: Murine retina fragments were embedded in fibrin gel and incubated for 7 days in the presence of VEGF (75 ng/mL) and increasing concentrations of K5-N,OS(H). At day 7, endothelial sprouts invading the surrounding fibrin matrix were counted. Data are the mean ± SEM of four independent determinations. F: Chick embryo CAMs were implanted at day 11 with alginate beads containing 100 ng/pellet of VEGF and increasing amounts of K5-N,OS(H). After 3 days, newly formed blood vessels converging toward the implant were counted under a stereo microscope. Data are the mean ± SEM of eight implants per experimental point. G: Elvax-40 pellets containing VEGF alone (200 ng) or VEGF plus 100 ng K5-N,OS(H) were implanted in the cornea of rabbit eyes. The angiogenic score was evaluated at day 11. Data are the mean ± SEM of three implants per experimental point. Insetsdg: Representative images of the inhibitory effect exerted by K5-N,OS(H) on the angiogenic response elicited by VEGF in the corresponding assays. Dashed circle, Elvax implant; dashed line, corneal limbus. *P < 0.05 and **P < 0.01 vs. vehicle (ANOVA).

Figure 2

K5-N,OS(H) inhibits the angiogenic activity of VEGF. A: HUVECs were incubated for 10 min at room temperature in the absence (control) or presence of 5.0 ng/mL VEGF and increasing concentrations of K5-N,OS(H), lysed, and probed in a Western blot with anti-pVEGFR2 (Y1175) antibody. Uniform loading of the gel was confirmed using an antifocal adhesion kinase (FAK) antibody. B: HUVECs were grown with 10 ng/mL VEGF in the absence (black bars) or presence (white bars) of 10 ng/mL K5-N,OS(H) and counted after 24 h. Cell proliferation was expressed as fold increase in respect to the control. Data are the mean ± SEM of three independent determinations. C: HUVEC spheroids were embedded in fibrin gel and incubated with 30 ng/mL VEGF in the absence (black bars) or presence (white bars) of 10 ng/mL K5-N,OS(H). Formation of radially growing cell sprouts was evaluated after 24 h of incubation. Data are the mean ± SEM of three independent determinations. D: Murine aortic rings were embedded in fibrin gel and incubated in the presence of VEGF (30 ng/mL) and increasing concentrations of K5-N,OS(H). After 5 days, vessel sprouts were counted under a stereo microscope. Data are the mean ± SEM of three independent determinations. E: Murine retina fragments were embedded in fibrin gel and incubated for 7 days in the presence of VEGF (75 ng/mL) and increasing concentrations of K5-N,OS(H). At day 7, endothelial sprouts invading the surrounding fibrin matrix were counted. Data are the mean ± SEM of four independent determinations. F: Chick embryo CAMs were implanted at day 11 with alginate beads containing 100 ng/pellet of VEGF and increasing amounts of K5-N,OS(H). After 3 days, newly formed blood vessels converging toward the implant were counted under a stereo microscope. Data are the mean ± SEM of eight implants per experimental point. G: Elvax-40 pellets containing VEGF alone (200 ng) or VEGF plus 100 ng K5-N,OS(H) were implanted in the cornea of rabbit eyes. The angiogenic score was evaluated at day 11. Data are the mean ± SEM of three implants per experimental point. Insetsdg: Representative images of the inhibitory effect exerted by K5-N,OS(H) on the angiogenic response elicited by VEGF in the corresponding assays. Dashed circle, Elvax implant; dashed line, corneal limbus. *P < 0.05 and **P < 0.01 vs. vehicle (ANOVA).

Close modal

Next, K5-N,OS(H) was tested in the chick embryo CAM and rabbit cornea assays. In the first assay, alginate beads adsorbed with 100 ng VEGF and increasing concentrations of K5-N,OS(H) were implanted on the CAM of 11-day-old chick embryos (30). As shown in Fig. 2F, K5-N,OS(H) caused a significant inhibition of VEGF-induced angiogenesis in the absence of any effect on embryonic development and survival. In the rabbit cornea assay, pellets of polymer Elvax-40 containing 200 ng VEGF with or without 100 ng K5-N,OS(H) were implanted in the cornea and the angiogenic score was evaluated 11 days thereafter (40). K5-N,OS(H) significantly inhibited VEGF-induced neovascularization also in this assay (Fig. 2G).

K5-N,OS(H) Inhibits Retinal Neovascularization in the Murine OIR Model

The effect of K5-N,OS(H) on retinal angiogenesis was evaluated in vivo in a murine OIR model in which VEGFR2 is the main mediator of the proangiogenic activity of hypoxia-upregulated VEGF (41). As shown in Fig. 3, intravitreal injection of increasing concentrations of K5-N,OS(H) (ranging from 0.06 to 60 ng/µL) caused a significant decrease of VEGFR2 phosphorylation in the retinas of PD17 OIR mice with no effect on the total levels of VEGFR2.

Figure 3

K5-N,OS(H) reduces VEGFR2 phosphorylation in the murine OIR model. OIR mice were intravitreally injected in the left eye at PD12 and PD15 with increasing concentrations of K5-N,OS(H) (ranging from 0.06 to 60 ng/μL). Right eyes were injected with vehicle. Retinas were explanted at PD17 and analyzed by Western blotting with anti-VEGFR2 and anti-pVEGFR2 antibodies. A: Representative blots depicting retinal levels of VEGFR2 and pVEGFR2 after treatment with vehicle or K5-N,OS(H). Uniform loading of the gel was confirmed using an anti–β-actin antibody. B: Densitometric analysis of pVEGFR2 levels normalized to VEGFR2 expression. Data are the mean ± SEM of five independent samples. *P < 0.05 and **P < 0.01 vs. vehicle; §P < 0.05 vs. 0.6 ng/μL K5-N,OS(H) (ANOVA).

Figure 3

K5-N,OS(H) reduces VEGFR2 phosphorylation in the murine OIR model. OIR mice were intravitreally injected in the left eye at PD12 and PD15 with increasing concentrations of K5-N,OS(H) (ranging from 0.06 to 60 ng/μL). Right eyes were injected with vehicle. Retinas were explanted at PD17 and analyzed by Western blotting with anti-VEGFR2 and anti-pVEGFR2 antibodies. A: Representative blots depicting retinal levels of VEGFR2 and pVEGFR2 after treatment with vehicle or K5-N,OS(H). Uniform loading of the gel was confirmed using an anti–β-actin antibody. B: Densitometric analysis of pVEGFR2 levels normalized to VEGFR2 expression. Data are the mean ± SEM of five independent samples. *P < 0.05 and **P < 0.01 vs. vehicle; §P < 0.05 vs. 0.6 ng/μL K5-N,OS(H) (ANOVA).

Close modal

On this basis, retinal vascularization was evaluated by CD31 immunohistochemistry after intravitreal administration of vehicle or K5-N,OS(H) in OIR mice. When compared with normoxic retinas (Fig. 4A), PD17 retinas of untreated and vehicle-treated OIR mice show the formation of a large avascular area in their central region and a prominent vessel regrowth leading to preretinal neovascular tufts in the midperipheral retina (Fig. 4B and C). In K5-N,OS(H)–treated eyes (Fig. 4D–G), the extent of avascular area was similar to those observed in vehicle-treated retinas (data not shown). However, the vessel tuft area appeared to be reduced with no preferential regional distribution. Quantitative analysis of CD31 immunoreactivity confirmed a significant dose-dependent decrease of the neovascular tuft area in K5-N,OS(H)–treated OIR retinas (Fig. 4H). Also, in keeping with its anti-inflammatory activity (4244), K5-N,OS(H) caused downregulation of the expression of various markers of inflammation in OIR retinas (Supplementary Fig. 1), with no significant changes in the expression of angiogenic growth factors known to play a role in OIR, including VEGF (Supplementary Fig. 2), and in the amount of apoptosis-related proteins, including Bax, Bcl-2, and cytochrome c (Supplementary Fig. 3).

Figure 4

K5-N,OS(H) inhibits retinal neovascularization in the murine OIR model. CD31 immunostaining of flat-mounted retinas from mice exposed to room air (A) or to 75 ± 2% oxygen from PD7 to PD12 and left untreated (B) or intravitreally injected with vehicle (C) or with increasing concentrations of K5-N,OS(H) in the left eye at PD12 and PD15 (DG). Right eyes were injected with vehicle. Retinas were explanted at PD17. Hyperoxia followed by normoxia for 5 days produced the central loss of blood vessels and the formation of tufts in the midperipheral retina. K5-N,OS(H) reduced drastically the vessel tuft area (arrows). The retina shown in C is contralateral to the K5-N,OS(H)–treated retina shown in G. Scale bar, 1 mm. H: Quantitative analysis of the total area of preretinal neovascular tufts was performed on the entire retina by dividing the retina in four quadrants (I–IV, white boundaries). Data are the mean ± SEM from five retinas. *P < 0.05 and **P < 0.01 vs. vehicle (ANOVA).

Figure 4

K5-N,OS(H) inhibits retinal neovascularization in the murine OIR model. CD31 immunostaining of flat-mounted retinas from mice exposed to room air (A) or to 75 ± 2% oxygen from PD7 to PD12 and left untreated (B) or intravitreally injected with vehicle (C) or with increasing concentrations of K5-N,OS(H) in the left eye at PD12 and PD15 (DG). Right eyes were injected with vehicle. Retinas were explanted at PD17. Hyperoxia followed by normoxia for 5 days produced the central loss of blood vessels and the formation of tufts in the midperipheral retina. K5-N,OS(H) reduced drastically the vessel tuft area (arrows). The retina shown in C is contralateral to the K5-N,OS(H)–treated retina shown in G. Scale bar, 1 mm. H: Quantitative analysis of the total area of preretinal neovascular tufts was performed on the entire retina by dividing the retina in four quadrants (I–IV, white boundaries). Data are the mean ± SEM from five retinas. *P < 0.05 and **P < 0.01 vs. vehicle (ANOVA).

Close modal

K5-N,OS(H) Is a Multitarget Binder for Heparin-Binding Angiogenic Factors

Various heparin-binding proinflammatory/angiogenic factors other than VEGF play an important role in PDR, including FGF2, IGF-1, IL8, PDGF-B, SDF1α, CTGF, HGF, HMGB1, and placental growth factor-2 (PlGF2) (25). When tested by SPR analysis, these proteins bind immobilized heparin with Kd values ranging between 10 and 600 nmol/L. Notably, K5-N,OS(H) caused a dose-dependent inhibition of the binding of all the factors tested to the heparin sensor chip, with ID50 values ranging between 0.01 and 0.2 µg/mL (Fig. 5A). Of note, no correlation exists between the capacity of K5N,OS(H) to hamper the interaction of the various proteins with immobilized heparin (expressed as ID50 value) and their affinity for the immobilized GAG (expressed as Kd value) (Fig. 5B).

Figure 5

K5-N,OS(H) inhibits the binding of PDR-related angiogenic growth factors to immobilized heparin. A: VEGF (black circles), SDF1α (white circles), FGF2 (black triangles), IL8 (white triangles), HGF (black squares), HMGB1 (white squares), PlGF2 (black diamonds), PDGF-B (white diamonds), and CTGF (black down-pointing triangles) were coinjected with increasing concentrations of K5-N,OS(H) on heparin immobilized to a Biacore sensor chip. The response (in RU) was recorded at the end of injection and plotted as a function of K5-N,OS(H) concentration. B: For each protein, its affinity for heparin interaction (expressed as SPR-assessed Kd value) was plotted vs. the ID50 value of K5-N,OS(H)–dependent inhibition of its binding to immobilized heparin. No significant correlation was observed between the two parameters. C: Wounded HUVEC monolayers were treated with 50 ng/mL of the indicated angiogenic factor in the absence or presence of 100 ng/mL K5-N,OS(H). After 24 h, HUVECs invading the wound were quantified by computerized analysis of the digitalized images. Data are the mean ± SEM of six measurements per sample. All the angiogenic factors exerted a significant stimulation of wound repair in respect to untreated wounded monolayers (P < 0.05 or better). This stimulation was inhibited by K5-N,OS(H) that was ineffective on control cells (*P < 0.05 and **P < 0.01 vs. angiogenic factor alone, Student t test).

Figure 5

K5-N,OS(H) inhibits the binding of PDR-related angiogenic growth factors to immobilized heparin. A: VEGF (black circles), SDF1α (white circles), FGF2 (black triangles), IL8 (white triangles), HGF (black squares), HMGB1 (white squares), PlGF2 (black diamonds), PDGF-B (white diamonds), and CTGF (black down-pointing triangles) were coinjected with increasing concentrations of K5-N,OS(H) on heparin immobilized to a Biacore sensor chip. The response (in RU) was recorded at the end of injection and plotted as a function of K5-N,OS(H) concentration. B: For each protein, its affinity for heparin interaction (expressed as SPR-assessed Kd value) was plotted vs. the ID50 value of K5-N,OS(H)–dependent inhibition of its binding to immobilized heparin. No significant correlation was observed between the two parameters. C: Wounded HUVEC monolayers were treated with 50 ng/mL of the indicated angiogenic factor in the absence or presence of 100 ng/mL K5-N,OS(H). After 24 h, HUVECs invading the wound were quantified by computerized analysis of the digitalized images. Data are the mean ± SEM of six measurements per sample. All the angiogenic factors exerted a significant stimulation of wound repair in respect to untreated wounded monolayers (P < 0.05 or better). This stimulation was inhibited by K5-N,OS(H) that was ineffective on control cells (*P < 0.05 and **P < 0.01 vs. angiogenic factor alone, Student t test).

Close modal

In keeping with its binding capacity, K5N,OS(H) inhibits the proliferative/motogenic response elicited by the various heparin-binding angiogenic factors on mechanically wounded HUVEC monolayers (Fig. 5C). Thus, K5-N,OS(H) acts as a multitarget antagonist for a variety of heparin-binding proinflammatory/angiogenic factors involved in PDR.

K5-N,OS(H) Inhibits the Angiogenic Activity of Vitreous Fluid From Patients With PDR

The evaluation of the angiogenic effects exerted by the vitreous humor obtained after pars plana vitrectomy may provide a useful tool for a preclinical screening of angiostatic molecules with potential implications for the therapy of PDR (6,4547). To this aim, preliminary experiments were performed to define the amount of PDR vitreous fluid that exerts an optimal angiogenic response in the HUVEC sprouting and chick embryo CAM assays. Given the limited amount of fluid available from each patient, samples obtained from five to eight patients were pooled together (Table 1). The results showed that a 1:4 (vol:vol) dilution of PDR vitreous fluid in cell culture medium provided an angiogenic stimulus in the HUVEC sprouting assay similar to that exerted by 30 ng/mL VEGF and that 2.0 µL/pellet of the same sample induce neovessel formation in the CAM in a manner similar to 100 ng/pellet of VEGF. Comparable results were obtained with three distinct pools from different patients with PDR (data not shown), whereas no angiogenic response was elicited by a pool of vitreous fluid samples from macular hole patients (Supplementary Fig. 4). As shown in Fig. 6A and B, K5-N,OS(H) hampered the proangiogenic activity exerted by pools of PDR vitreous fluid both in the HUVEC sprouting assay and in the CAM assay with ID50 values equal to 1.0 ng/mL and 0.1 μg/pellet, respectively.

Figure 6

K5-N,OS(H) inhibits the angiogenic activity of vitreous fluid from patients with PDR. A: HUVEC spheroids embedded in fibrin gel were incubated with a pool of PDR vitreous fluid samples (1:4 dilution in cell culture medium) in the presence of increasing concentrations of K5-N,OS(H). Formation of radially growing sprouts was evaluated after 24 h of incubation. Data are the mean ± SEM of three independent determinations. B: CAMs were implanted at day 11 with alginate beads containing 2 µL of pooled PDR vitreous fluid and increasing concentrations of K5-N,OS(H). After 3 days, newly formed blood vessels converging toward the implant were counted under a stereo microscope. Data are the mean ± SEM of eight implants per experimental point. *P < 0.05 and **P < 0.01 vs. control (ANOVA). C: HUVEC spheroids were incubated with a 1:4 dilution of individual vitreous fluid samples obtained from 10 patients (P1–P10) with PDR in the absence (black bars) or in the presence of 10 ng/mL K5-N,OS(H) (white bars) or 150 μg/mL bevacizumab (dashed bars). Formation of radially growing sprouts was evaluated after 24 h of incubation, and data were normalized to the activity exerted in the same assay by 30 ng/mL VEGF, whose value was set equal to 1 and used as an internal standard in all the experiments. Data are the mean ± SEM of 40 spheroids per experimental point. D: The graph compares the inhibitory effects exerted by bevacizumab (black circles) and K5-N,OS(H) (white circles) on the angiogenic activity exerted by each individual vitreous fluid in the HUVEC spheroid assay as evaluated in panel C. K5-N,OS(H) appears to be more effective in most of the samples tested. *P < 0.05 and **P < 0.01 vs. untreated vitreous fluid (ANOVA). E: Angiogenic activity of untreated, bevacizumab-treated, and K5-N,OS(H)–treated vitreous fluid samples from patients with PDR that did (open bars) or did not (black bars) undergo intravitreal treatment with VEGF blockers 10–15 days prior to vitrectomy. Data, normalized to the activity of VEGF in the same assay, are the mean ± SEM of five patients per group. ns, not statistically significant. *P < 0.05.

Figure 6

K5-N,OS(H) inhibits the angiogenic activity of vitreous fluid from patients with PDR. A: HUVEC spheroids embedded in fibrin gel were incubated with a pool of PDR vitreous fluid samples (1:4 dilution in cell culture medium) in the presence of increasing concentrations of K5-N,OS(H). Formation of radially growing sprouts was evaluated after 24 h of incubation. Data are the mean ± SEM of three independent determinations. B: CAMs were implanted at day 11 with alginate beads containing 2 µL of pooled PDR vitreous fluid and increasing concentrations of K5-N,OS(H). After 3 days, newly formed blood vessels converging toward the implant were counted under a stereo microscope. Data are the mean ± SEM of eight implants per experimental point. *P < 0.05 and **P < 0.01 vs. control (ANOVA). C: HUVEC spheroids were incubated with a 1:4 dilution of individual vitreous fluid samples obtained from 10 patients (P1–P10) with PDR in the absence (black bars) or in the presence of 10 ng/mL K5-N,OS(H) (white bars) or 150 μg/mL bevacizumab (dashed bars). Formation of radially growing sprouts was evaluated after 24 h of incubation, and data were normalized to the activity exerted in the same assay by 30 ng/mL VEGF, whose value was set equal to 1 and used as an internal standard in all the experiments. Data are the mean ± SEM of 40 spheroids per experimental point. D: The graph compares the inhibitory effects exerted by bevacizumab (black circles) and K5-N,OS(H) (white circles) on the angiogenic activity exerted by each individual vitreous fluid in the HUVEC spheroid assay as evaluated in panel C. K5-N,OS(H) appears to be more effective in most of the samples tested. *P < 0.05 and **P < 0.01 vs. untreated vitreous fluid (ANOVA). E: Angiogenic activity of untreated, bevacizumab-treated, and K5-N,OS(H)–treated vitreous fluid samples from patients with PDR that did (open bars) or did not (black bars) undergo intravitreal treatment with VEGF blockers 10–15 days prior to vitrectomy. Data, normalized to the activity of VEGF in the same assay, are the mean ± SEM of five patients per group. ns, not statistically significant. *P < 0.05.

Close modal

On this basis, vitreous fluids from 10 patients with PDR (Table 1) were individually tested in the HUVEC sprouting assay with or without 10 ng/mL K5-N,OS(H) (corresponding to 0.7 nmol/L final concentration) or 150 μg/mL bevacizumab (corresponding to 1.0 µmol/L final concentration). As shown in Fig. 6C, PDR vitreous fluid samples exert a different angiogenic response when their activity was normalized to that exerted by 30 ng/mL VEGF in the same assay. In 8 out of the 10 patients tested, K5-N,OS(H) caused a significant inhibition of the angiogenic activity of PDR vitreous fluid whereas bevacizumab was effective on three samples only, with an average percent of inhibition equal to 69 ± 8 and 33 ± 7% (P < 0.01) for the two compounds, respectively (Fig. 6C and D). Accordingly, the inhibitory action of K5-N,OS(H) was significantly more potent than bevacizumab in eight vitreous fluid samples (Fig. 6D). For both inhibitors, their efficacy was not related to the angiogenic potency of the PDR vitreous fluid tested nor to the clinical features of the enrolled patients with PDR (data not shown), including intravitreal pretreatment with VEGF blockers (Fig. 6E).

Here, we identified K5-N,OS(H) as a biotechnological HS-like molecule highly effective in inhibiting the activity of VEGF in different angiogenesis assays, including a murine model of OIR. Of note, K5-N,OS(H) also inhibits the angiogenic responses triggered by human vitreous fluid samples collected from patients with PDR, being more effective than the specific VEGF blocker bevacizumab. The remarkable efficacy of K5-N,OS(H) may be related to its capacity to act as a multitarget antagonist for a variety of heparin-binding proangiogenic/proinflammatory factors upregulated in PDR vitreous humor.

K5-N,OS(H) is a highly N,O-sulfated K5 derivative (28) with antiangiogenic, antineoplastic, and antiviral activities (26,27) but endowed with negligible anticoagulant activity when compared with heparin (26). Also, K5-N,OS(H) inhibits proinflammatory cytokine production by mononuclear cells (44). Accordingly, systemic administration of K5-N,OS(H) at doses as high as 1.0 mg/kg body weight exerts an anti-inflammatory activity in different models of ischemia/reperfusion injury in the absence of toxic effects (42,43).

Previous observations had shown the capacity of K5-N,OS(H) to bind FGF2 and HIV-Tat, thus inhibiting their angiogenic activity (2628). Here, K5-N,OS(H) also binds with high affinity the major heparin-binding VEGF-A165 isoform expressed in PDR (3). Indeed, given that VEGF-A165 interaction with immobilized heparin occurs with a Kd value equal to 100 nmol/L (38), the competition experiments shown in Fig. 1C demonstrate that VEGF/K5-N,OS(H) interaction occurs with an 80-fold higher affinity, leading to a Kd value equal to ∼1.3 nmol/L. Accordingly, K5-N,OS(H) is more potent than unfractionated heparin in hampering the binding of VEGF to immobilized sVEGFR2 and in preventing the formation of HSPG/VEGF/VEGFR2 ternary complexes. Furthermore, in all these assays, K5-N,OS(H) was more active than the low sulfated derivatives K5-N,OS(L) and K5-NS whereas K5 was inactive (Table 1). Thus, the capacity of K5 derivatives to bind VEGF appears to depend on the backbone structure, charge distribution, and degree of sulfation of the GAG chain.

In keeping with these observations, K5-N,OS(H) inhibits different steps of the angiogenic process mediated by VEGF in vitro, ex vivo, and in vivo. Remarkably, intravitreal injection of K5-N,OS(H) also reduced the formation of preretinal neovascular tufts in a murine OIR model in which VEGFR2 mediates the proangiogenic activity of hypoxia-upregulated VEGF (41). In keeping with the aforementioned anti-inflammatory potential of K5-N,OS(H), this was paralleled by a significant downregulation of proinflammatory mediators in OIR retinas, with no effect on the expression of various angiogenic factors known to play a role in OIR, including VEGF. Of note, K5-N,OS(H) did not affect retinal health, as shown by the analysis of the levels of the apoptotic markers Bax, Bcl-2, and cytochrome c.

The OIR model recapitulates various aspects of proliferative retinopathies, including PDR (23). Indeed, although the metabolic changes that characterize PDR are not present in OIR mice, many features of human PDR are observed in the OIR model in which early vessel loss leads to neovascularization with consequent blood-retinal barrier breakdown and neuronal damage (23). K5-N,OS(H) inhibits the formation of preretinal neovascular tufts without affecting the extent of the avascular area in OIR retinas. The mechanisms that control the balance between pathological and healthy angiogenesis in the OIR model are not fully elucidated (47). Further experiments will be required to assess the impact of K5-N,OS(H) on healthy retinal regeneration.

The inhibitory effect of injected K5-N,OS(H) in the OIR model occurred at concentrations as low as 0.6 ng/μL (corresponding to an intravitreal concentration equal to 7.0 nmol/L when considering a vitreous volume of about 5.0 µL [48]) and was paralleled by a significant inhibition of VEGFR2 phosphorylation in OIR retinas, thus confirming the capacity of K5-N,OS(H) to act in vivo as a VEGF antagonist in the retinal microenvironment. Relevant to this point, previous observations had shown that intravitreal injection of heparin/HS or of a synthetic heparin analog reduces the angioproliferative changes in the OIR model (20,21). However, the effect occurred at intravitreal concentrations (equal to 1.0 and 10 mmol/L, respectively) >100,000-fold higher than the minimal K5-N,OS(H) effective dose.

The assessment of the angiogenic potential of the PDR vitreous humor may provide a useful tool for a preclinical evaluation of therapeutic angiostatic molecules (6,4547). Here, PDR vitreous fluid elicits a potent angiogenic response in vitro and in vivo. Interestingly, significant differences in angiogenic potency were observed among the individual vitreous samples tested. These differences were not related to any of the clinical features of the enrolled patients with PDR (listed in Table 1), including intravitreal treatment with VEGF blockers performed 10–15 days prior to vitrectomy, presence of vitreous hemorrhage, or macular edema (Fig. 6E and data not shown). As anticipated, K5-N,OS(H) hampers the angiogenic activity exerted by PDR vitreous fluid pooled from different patients. Also, when tested on individual samples, the inhibitory effect of K5-N,OS(H) was more potent than that exerted by bevacizumab in 8 out of 10 patients with PDR that were examined. This occurred despite the fact that K5-N,OS(H) was tested at a final molar concentration 1,000-fold lower than the anti-VEGF antibody. Interestingly, we did not observe a significant relationship between the inhibitory effect exerted by bevacizumab or K5-N,OS(H) and the clinical features of patients with PDR, including intravitreal pretreatment with VEGF blockers (Fig. 6E). Further studies on a larger cohort of patients will be required to confirm these observations.

The remarkable efficacy shown by K5-N,OS(H) in reducing the angiogenic responses in the murine OIR model and in the endothelium treated with PDR vitreous fluid may be related to its capacity to bind and inhibit the biological activity of a variety of heparin-binding proinflammatory/angiogenic factors besides VEGF, including FGF2, IGF-1, IL8, PlGF2, PDGF-B, SDF1α, CTGF, HGF, and HMGB1, all present in the vitreous humor from patients with PDR (25). Of note, the potency of the K5-N,OS(H) interaction was unrelated to the affinity of these proteins for unfractionated heparin, indicating that distinct molecular determinants (e.g., differences in backbone structure and charge distribution) mediate their binding to the two GAGs. The possibility that the overall angiogenic potential of PDR vitreous humor may represent the result of the synergistic action of the various angiogenic factors is supported by the observation that the amount of recombinant VEGF required to induce in our assays an angiogenic response similar to that exerted by the pooled PDR vitreous fluid far exceeds the concentration of the growth factor detectable in PDR vitreous humor (median VEGF levels equal to 345 pg/mL [45]). The better performance of K5-N,OS(H) versus bevacizumab in inhibiting the angiogenic activity of PDR vitreous fluid substantiates this hypothesis.

In conclusion, our data provide compelling experimental evidence that K5-N,OS(H) represents a novel multitarget molecule with potential implications for more efficacious therapeutic interventions aimed at inhibiting the neovascular responses that occur in the retina of patients with PDR.

Acknowledgments. The authors thank I. Fornaciari (University of Pisa) for technical assistance and D. Rusciano (BIOOS Italia), M. Ziche (University of Siena), and G. Casini (University of Pisa) for helpful discussion and criticisms. CHO-K1 and A745 CHO cells were provided by J.D. Esko (University of California, La Jolla, CA). Human recombinant VEGF and pcDNA3 harboring the ECD-VEGFR2-YFP cDNA were provided by K. Ballmer-Hofer, and PlGF2 was provided by S. De Falco (IGB, Naples, Italy).

Funding. This work was supported in part by grants from Istituto Toscano Tumori to L.M., Regione Toscana to P.B., Ministero dell’Istruzione, Università e Ricerca (FIRB project RBAP11H2R9 2011), and Associazione Italiana per la Ricerca sul Cancro (AIRC grant 14395) to M.P., and BIOOS Italia to L.M., P.B., and M.P.

Duality of Interest. P.O. is an employee of Glycores 2000 and inventor of the patent EP1366082 and its international counterparts. No other potential conflicts of interest relevant to this article were reported.

Author Contributions. S.R. designed and performed experiments, analyzed data, and wrote the manuscript. M.D.M., M.B., A.B., P.C., M.Co., M.Ca., A.C., L.M., and P.O. designed and performed experiments and analyzed data. P.B. and F.S. analyzed data, contributed to discussion, reviewed the manuscript, and secured funding. M.P. supervised the project, designed experiments, analyzed data, wrote the manuscript, and secured funding. M.P. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Wild
S
,
Roglic
G
,
Green
A
,
Sicree
R
,
King
H
.
Global prevalence of diabetes: estimates for the year 2000 and projections for 2030
.
Diabetes Care
2004
;
27
:
1047
1053
[PubMed]
2.
Gariano
RF
,
Gardner
TW
.
Retinal angiogenesis in development and disease
.
Nature
2005
;
438
:
960
966
[PubMed]
3.
Simó
R
,
Carrasco
E
,
García-Ramírez
M
,
Hernández
C
.
Angiogenic and antiangiogenic factors in proliferative diabetic retinopathy
.
Curr Diabetes Rev
2006
;
2
:
71
98
[PubMed]
4.
Sherris
D
.
Ocular drug development—future directions
.
Angiogenesis
2007
;
10
:
71
76
[PubMed]
5.
Wang
S
,
Park
JK
,
Duh
EJ
.
Novel targets against retinal angiogenesis in diabetic retinopathy
.
Curr Diab Rep
2012
;
12
:
355
363
[PubMed]
6.
Aiello
LP
,
Avery
RL
,
Arrigg
PG
, et al
.
Vascular endothelial growth factor in ocular fluid of patients with diabetic retinopathy and other retinal disorders
.
N Engl J Med
1994
;
331
:
1480
1487
[PubMed]
7.
Tang
J
,
Kern
TS
.
Inflammation in diabetic retinopathy
.
Prog Retin Eye Res
2011
;
30
:
343
358
[PubMed]
8.
Mathews
MK
,
Merges
C
,
McLeod
DS
,
Lutty
GA
.
Vascular endothelial growth factor and vascular permeability changes in human diabetic retinopathy
.
Invest Ophthalmol Vis Sci
1997
;
38
:
2729
2741
[PubMed]
9.
Adamis
AP
,
Shima
DT
.
The role of vascular endothelial growth factor in ocular health and disease
.
Retina
2005
;
25
:
111
118
[PubMed]
10.
Penn
JS
,
Madan
A
,
Caldwell
RB
,
Bartoli
M
,
Caldwell
RW
,
Hartnett
ME
.
Vascular endothelial growth factor in eye disease
.
Prog Retin Eye Res
2008
;
27
:
331
371
[PubMed]
11.
Nicholson
BP
,
Schachat
AP
.
A review of clinical trials of anti-VEGF agents for diabetic retinopathy
.
Graefes Arch Clin Exp Ophthalmol
2010
;
248
:
915
930
[PubMed]
12.
Boyer
DS
,
Hopkins
JJ
,
Sorof
J
,
Ehrlich
JS
.
Anti-vascular endothelial growth factor therapy for diabetic macular edema
.
Ther Adv Endocrinol Metab
2013
;
4
:
151
169
[PubMed]
13.
Falavarjani
KG
,
Nguyen
QD
.
Adverse events and complications associated with intravitreal injection of anti-VEGF agents: a review of literature
.
Eye (Lond)
2013
;
27
:
787
794
[PubMed]
14.
Cheung
N
,
Wong
IY
,
Wong
TY
.
Ocular anti-VEGF therapy for diabetic retinopathy: overview of clinical efficacy and evolving applications
.
Diabetes Care
2014
;
37
:
900
905
[PubMed]
15.
Chen
Y
,
Hu
Y
,
Lin
M
, et al
.
Therapeutic effects of PPARα agonists on diabetic retinopathy in type 1 diabetes models
.
Diabetes
2013
;
62
:
261
272
[PubMed]
16.
Lee
K
,
Hu
Y
,
Ding
L
, et al
.
Therapeutic potential of a monoclonal antibody blocking the Wnt pathway in diabetic retinopathy
.
Diabetes
2012
;
61
:
2948
2957
[PubMed]
17.
Rusnati
M
,
Presta
M
.
Interaction of angiogenic basic fibroblast growth factor with endothelial cell heparan sulfate proteoglycans. Biological implications in neovascularization
.
Int J Clin Lab Res
1996
;
26
:
15
23
[PubMed]
18.
Grünewald FS, Prota AE, Giese A, Ballmer-Hofer K. Structure-function analysis of VEGF receptor activation and the role of coreceptors in angiogenic signaling. Biochim Biophys Acta 2010;1804:567–580
19.
Presta
M
,
Leali
D
,
Stabile
H
, et al
.
Heparin derivatives as angiogenesis inhibitors
.
Curr Pharm Des
2003
;
9
:
553
566
[PubMed]
20.
Lange
C
,
Ehlken
C
,
Martin
G
, et al
.
Intravitreal injection of the heparin analog 5-amino-2-naphthalenesulfonate reduces retinal neovascularization in mice
.
Exp Eye Res
2007
;
85
:
323
327
[PubMed]
21.
Nishiguchi
KM
,
Kataoka
K
,
Kachi
S
,
Komeima
K
,
Terasaki
H
.
Regulation of pathologic retinal angiogenesis in mice and inhibition of VEGF-VEGFR2 binding by soluble heparan sulfate
.
PLoS ONE
2010
;
5
:
e13493
[PubMed]
22.
Smith
LE
,
Wesolowski
E
,
McLellan
A
, et al
.
Oxygen-induced retinopathy in the mouse
.
Invest Ophthalmol Vis Sci
1994
;
35
:
101
111
[PubMed]
23.
Stahl
A
,
Connor
KM
,
Sapieha
P
, et al
.
The mouse retina as an angiogenesis model
.
Invest Ophthalmol Vis Sci
2010
;
51
:
2813
2826
[PubMed]
24.
Zhang
D
,
Kaufman
PL
,
Gao
G
,
Saunders
RA
,
Ma
JX
.
Intravitreal injection of plasminogen kringle 5, an endogenous angiogenic inhibitor, arrests retinal neovascularization in rats
.
Diabetologia
2001
;
44
:
757
765
[PubMed]
25.
Vann
WF
,
Schmidt
MA
,
Jann
B
,
Jann
K
.
The structure of the capsular polysaccharide (K5 antigen) of urinary-tract-infective Escherichia coli 010:K5:H4. A polymer similar to desulfo-heparin
.
Eur J Biochem
1981
;
116
:
359
364
[PubMed]
26.
Rusnati
M
,
Oreste
P
,
Zoppetti
G
,
Presta
M
.
Biotechnological engineering of heparin/heparan sulphate: a novel area of multi-target drug discovery
.
Curr Pharm Des
2005
;
11
:
2489
2499
[PubMed]
27.
Rusnati
M
,
Urbinati
C
.
Polysulfated/sulfonated compounds for the development of drugs at the crossroad of viral infection and oncogenesis
.
Curr Pharm Des
2009
;
15
:
2946
2957
[PubMed]
28.
Leali
D
,
Belleri
M
,
Urbinati
C
, et al
.
Fibroblast growth factor-2 antagonist activity and angiostatic capacity of sulfated Escherichia coli K5 polysaccharide derivatives
.
J Biol Chem
2001
;
276
:
37900
37908
[PubMed]
29.
Bugatti
A
,
Giagulli
C
,
Urbinati
C
, et al
.
Molecular interaction studies of HIV-1 matrix protein p17 and heparin: identification of the heparin-binding motif of p17 as a target for the development of multitarget antagonists
.
J Biol Chem
2013
;
288
:
1150
1161
[PubMed]
30.
Mitola
S
,
Moroni
E
,
Ravelli
C
,
Andres
G
,
Belleri
M
,
Presta
M
.
Angiopoietin-1 mediates the proangiogenic activity of the bone morphogenic protein antagonist Drm
.
Blood
2008
;
112
:
1154
1157
[PubMed]
31.
Ronca
R
,
Benzoni
P
,
Leali
D
, et al
.
Antiangiogenic activity of a neutralizing human single-chain antibody fragment against fibroblast growth factor receptor 1
.
Mol Cancer Ther
2010
;
9
:
3244
3253
[PubMed]
32.
Rezzola
S
,
Belleri
M
,
Gariano
G
, et al
.
In vitro and ex vivo retina angiogenesis assays
.
Angiogenesis
2014
;17:429–442
[PubMed]
33.
Rezzola
S
,
Belleri
M
,
Ribatti
D
,
Costagliola
C
,
Presta
M
,
Semeraro
F
.
A novel ex vivo murine retina angiogenesis (EMRA) assay
.
Exp Eye Res
2013
;
112
:
51
56
[PubMed]
34.
Monti
M
,
Donnini
S
,
Morbidelli
L
, et al
.
PKCε activation promotes FGF-2 exocytosis and induces endothelial cell proliferation and sprouting
.
J Mol Cell Cardiol
2013
;
63
:
107
117
[PubMed]
35.
Dal Monte
M
,
Ristori
C
,
Cammalleri
M
,
Bagnoli
P
.
Effects of somatostatin analogues on retinal angiogenesis in a mouse model of oxygen-induced retinopathy: involvement of the somatostatin receptor subtype 2
.
Invest Ophthalmol Vis Sci
2009
;
50
:
3596
3606
[PubMed]
36.
Dal Monte
M
,
Casini
G
,
la Marca
G
,
Isacchi
B
,
Filippi
L
,
Bagnoli
P
.
Eye drop propranolol administration promotes the recovery of oxygen-induced retinopathy in mice
.
Exp Eye Res
2013
;
111
:
27
35
[PubMed]
37.
Zilberberg
L
,
Shinkaruk
S
,
Lequin
O
, et al
.
Structure and inhibitory effects on angiogenesis and tumor development of a new vascular endothelial growth inhibitor
.
J Biol Chem
2003
;
278
:
35564
35573
[PubMed]
38.
Pisano
C
,
Aulicino
C
,
Vesci
L
, et al
.
Undersulfated, low-molecular-weight glycol-split heparin as an antiangiogenic VEGF antagonist
.
Glycobiology
2005
;
15
:
1C
6C
[PubMed]
39.
Mitola
S
,
Ravelli
C
,
Moroni
E
, et al
.
Gremlin is a novel agonist of the major proangiogenic receptor VEGFR2
.
Blood
2010
;
116
:
3677
3680
[PubMed]
40.
Ziche
M
,
Morbidelli
L
.
The corneal pocket assay
.
Methods Mol Biol
2009
;
467
:
319
329
[PubMed]
41.
Witmer
AN
,
Vrensen
GF
,
Van Noorden
CJ
,
Schlingemann
RO
.
Vascular endothelial growth factors and angiogenesis in eye disease
.
Prog Retin Eye Res
2003
;
22
:
1
29
[PubMed]
42.
Collino
M
,
Pini
A
,
Mastroianni
R
, et al
.
The non-anticoagulant heparin-like K5 polysaccharide derivative K5-N,OSepi attenuates myocardial ischaemia/reperfusion injury
[published correction appears in J Cell Mol Med 2013;17:1652].
J Cell Mol Med
2012
;
16
:
2196
2207
[PubMed]
43.
Collino
M
,
Castiglia
S
,
Manoni
M
, et al
.
Effects of a semi-synthetic N-,O-sulfated glycosaminoglycan K5 polysaccharide derivative in a rat model of cerebral ischaemia/reperfusion injury
.
Thromb Haemost
2009
;
102
:
837
845
[PubMed]
44.
Gori
AM
,
Attanasio
M
,
Gazzini
A
, et al
.
Cytokine gene expression and production by human LPS-stimulated mononuclear cells are inhibited by sulfated heparin-like semi-synthetic derivatives
.
J Thromb Haemost
2004
;
2
:
1657
1662
[PubMed]
45.
Watanabe
D
,
Suzuma
K
,
Matsui
S
, et al
.
Erythropoietin as a retinal angiogenic factor in proliferative diabetic retinopathy
.
N Engl J Med
2005
;
353
:
782
792
[PubMed]
46.
Yang
J
,
Klassen
H
,
Pries
M
,
Wang
W
,
Nissen
MH
.
Vitreous humor and albumin augment the proliferation of cultured retinal precursor cells
.
J Neurosci Res
2009
;
87
:
495
502
[PubMed]
47.
Scott
A
,
Fruttiger
M
.
Oxygen-induced retinopathy: a model for vascular pathology in the retina
.
Eye (Lond)
2010
;
24
:
416
421
[PubMed]
48.
Remtulla
S
,
Hallett
PE
.
A schematic eye for the mouse, and comparisons with the rat
.
Vision Res
1985
;
25
:
21
31
[PubMed]

Supplementary data