Effect of subconjuctival and intraocular bevacizumab injection on angiogenic gene expression levels in a mouse model of corneal neovascularization

Olga Dratviman-Storobinsky, Bat-Chen R Avraham-Lubin, Murat Hasanreisoglu, Nitza Goldenberg-Cohen, Olga Dratviman-Storobinsky, Bat-Chen R Avraham-Lubin, Murat Hasanreisoglu, Nitza Goldenberg-Cohen

Abstract

Purpose: This study sought to characterize the expression of angiogenesis-related genes in a mouse model of corneal neovascularization, either untreated or after treatment with a single injection of bevacizumab by three different routes. In addition, the effectiveness of the treatment was compared to a rabbit model.

Methods: A chemical burn was induced in the mid-cornea of the right eye in 119 mice; 56 of them were untreated and 63 were bevacizumab-treated. Neovascularization was evaluated 2, 4, 8, 10, and 14 days later using digital photos, angiography and India ink perfusion. The relative area of new blood vessels was analyzed using slit-lamp examination in vivo and on histological and flat-mount sections. The levels of gene expression involved in the angiogenic process vascular endothelial growth factor [VEGF], insulin-like growth factor-1 [IGF-1], pigment epithelium derived factor [PEDF], and macrophage-inflammatory protein-2 [MIP-2]) were measured by a real-time polymerase chain reaction. Six rabbits underwent the same injury and treatment, and the response was compared to the mouse model.

Results: Neovascularization was first observed two days after injury. The affected section increased from 11.24% (+/-7.0) of the corneal area to 47.42% (+/-25.4) on day 8 and 50.62% (+/-24.7) on day 10. In the mice treated with bevacizumab, the relative area of neovascularization was significantly lower at the peak time points (p<0.005): 24.90% (+/-21.8) on day 8 and 28.29% (+/-20.9) on day 10. Spontaneous regression was observed on day 14 in both groups, to 26.98% (+/-19.9) in the untreated mice and 10.97% (+/-10.8) in the bevacizumab-treated mice (p<0.005). Rabbits also showed peak corneal neovascularization on days 8-10, with significant regression of the vessels following intracameral bevacizumab injection. In the mice, intraocular (intravitreal, intracameral) injection was more effective than subconjuctival injection. VEGF gene expression was upregulated in both the untreated and treated mice, but was slightly less in the treated mice. PEDF gene expression decreased in both the treated and untreated mice. In the untreated group, gene expression peaked (above baseline) at 14 days, and in the untreated mice, it had already peaked by day 8. IGF-1 was upregulated early in the model; at 8 days, there was only a slight change in the untreated group compared to a significant increase in the treated group. MIP-2 was upregulated in both groups in the early stage and returned to baseline on day 14.

Conclusions: Bevacizumab treatment partially inhibits the progressive corneal neovascularization induced by chemical damage in a mouse model. Treatment is more effective when administered via the intraocular than the subconjunctival route. The clinical findings are compatible with the angiographic and histologic data and are supported by molecular analysis showing a partial change in expression of proangiogenic genes. The molecular mechanisms involved in corneal neovascularization and inflammation warrant further exploration. These findings may have important therapeutic implications in the clinical setting.

Figures

Figure 1
Figure 1
Corneal neovascularization in rabbits. A-B: Clinical appearance of neovascularization in a rabbit cornea 10 days (peak) after chemical burn induction. Note the central scar (yellow) covering approximately 10% of the corneal area. C-D: Intracameral bevacizumab-treated cornea at 10 days. Note the reduced neovascularization.
Figure 2
Figure 2
Clinical appearance of neovascularization in a mouse cornea. Comparison between normal eye, without blood vessels on the cornea (A) and 8 days following cauterization without treatment, with high level of neovascularization (B). Less blood vessels observed after bevacizumab treatment by (H) intravitreal injection, (I) intracameral injection or (J) subconjunctival injection at the same time point. Fluorescein angiography 8 days after chemical burn induction showing (C) early leakage and (D-E) increased late leakage. Flat mount cornea of the control mouse showing transparent cornea without blood vessels (F). In the neovascular corneal model, studies with fluorescein dye (G) and India ink (K, black) reveal new vessels 8 days after induction of chemical burn, compared to the control avascular cornea (K), and a reduced level of NV detected following bevacizumab treatment (M-O).
Figure 3
Figure 3
Histology analysis using hematoxylin-eosin staining. No blood vessels detected in the normal cornea (A). Following cauterization, development of the scar (arrow) was observed in the epithelial and anterior stroma of the center cornea (B). New pathological blood vessels (C) were located in the superficial stroma, filled with erythrocytes (*).
Figure 4
Figure 4
Corneal flat-mount immunostaining for endothelial marker. Flat-mount corneas were stained with endothelial marker using anti-CD31 antibody. No positive staining detected in a control mouse (avascular cornea; A) compared to blood vessels detected in the flat mount cornea  8 days after cauterization (red staining; B).
Figure 5
Figure 5
Immunostaining for intraocular bevacizumab. No staining was detected using Anti-human antibody labeled with CY3 dye in an untreated mouse after chemical burn injury (A) without bevacizumab injection. Bevacizumab staining identified in the anterior chamber, filtrating into the cornea, following intraocular injection one day after injury (B).
Figure 6
Figure 6
Molecular analysis of gene expression levels after chemical cauterization in the untreated and bevacizumab treated mice. VEGF expression up-regulated in all time points, with a slight decrease following bevacizumab treatment (A). PEDF showed an inverse pattern of expression to VEGF (B). IGF-1 expression increased between days 8 and 10 following injury, with a higher level in the bevacizumab treated group (C). MIP-2 expression significantly increased in both the untreated and treated mice on day 2 and then dropped to near-normal levels by day 14, with higher level in the bevacizumab treated eyes (D).

References

    1. Cursiefen C, Chen L, Saint-Geniez M, Hamrah P, Jin Y, Rashid S, Pytowski B, Persaud K, Wu Y, Streilein JW, Dana R. Nonvascular VEGF receptor 3 expression by corneal epithelium maintains avascularity and vision. Proc Natl Acad Sci USA. 2006;103:11405–10.
    1. Cursiefen C, Rummelt C, Junemann A, Vorwerk C, Neuhuber W, Kruse FE, Schroedl F. Absence of blood and lymphatic vessels in the developing human cornea. Cornea. 2006;25:722–6.
    1. Bock F, Onderka J, Dietrich T, Bachmann B, Kruse FE, Paschke M, Zahn G, Cursiefen C. Bevacizumab as a potent inhibitor of inflammatory corneal angiogenesis and lymphangiogenesis. Invest Ophthalmol Vis Sci. 2007;48:2545–52.
    1. Samolov B, Steen B, Seregard S, van der Ploeg I, Montan P, Kvanta A. Delayed inflammation-associated corneal neovascularization in MMP-2-deficient mice. Exp Eye Res. 2005;80:159–66.
    1. Cursiefen C, Maruyama K, Jackson DG, Streilein JW, Kruse FE. Time course of angiogenesis and lymphangiogenesis after brief corneal inflammation. Cornea. 2006;25:443–7.
    1. Chang JH, Gabison EE, Kato T, Azar DT. Corneal neovascularization. Curr Opin Ophthalmol. 2001;12:242–9.
    1. Lee P, Wang CC, Adamis AP. Ocular neovascularization: an epidemiologic review. Surv Ophthalmol. 1998;43:245–69.
    1. Hashemian MN, Moghimi S, Kiumehr S, Riazi M, Amoli FA. Prevention and treatment of corneal neovascularization: comparison of different doses of subconjunctival bevacizumab with corticosteriod in experimental rats. Ophthalmic Res. 2009;42:90–5.
    1. Klebe S, Coster DJ, Williams KA. Rejection and acceptance of corneal allografts. Curr Opin Organ Transplant. 2009;14:4–9.
    1. Lafleur MA, Handsley MM, Edwards DR. Metalloproteinases and their inhibitors in angiogenesis. Expert Rev Mol Med. 2003;5:1–39.
    1. Segal E, Satchi-Fainaro R. Design and development of polymer conjugates as anti-angiogenic agents. Adv Drug Deliv Rev. 2009
    1. Metro G, Cappuzzo F. Emerging drugs for small-cell lung cancer. Expert Opin Emerg Drugs. 2009
    1. Hammady T, Rabanel JM, Dhanikula RS, Leclair G, Hildgen P. Functionalized nanospheres loaded with anti-angiogenic drugs: Cellular uptake and angiosuppressive efficacy. Eur J Pharm Biopharm. 2009;72:418–2719462478.
    1. Ferrara N, Kerbel RS. Angiogenesis as a therapeutic target. Nature. 2005;438:967–74.
    1. Penn JS, Madan A, Caldwell RB, Bartoli M, Caldwell RW, Hartnett ME. Vascular endothelial growth factor in eye disease. Prog Retin Eye Res. 2008;27:331–71.
    1. Phillips GD, Stone AM, Jones BD, Schultz JC, Whitehead RA, Knighton DR. Vascular endothelial growth factor (rhVEGF165) stimulates direct angiogenesis in the rabbit cornea. In Vivo. 1994;8:961–5.
    1. Gan L, Fagerholm P, Palmblad J. Vascular endothelial growth factor (VEGF) and its receptor VEGFR-2 in the regulation of corneal neovascularization and wound healing. Acta Ophthalmol Scand. 2004;82:557–63.
    1. Philipp W, Speicher L, Humpel C. Expression of vascular endothelial growth factor and its receptors in inflamed and vascularized human corneas. Invest Ophthalmol Vis Sci. 2000;41:2514–22.
    1. Iwasaki J, Nihira S. Anti-angiogenic therapy against gastrointestinal tract cancers. Jpn J Clin Oncol. 2009;39:543–51.
    1. Manzano RP, Peyman GA, Khan P, Carvounis PE, Kivilcim M, Ren M, Lake JC, Chevez-Barrios P. Inhibition of experimental corneal neovascularisation by bevacizumab (Avastin). Br J Ophthalmol. 2007;91:804–7.
    1. Papathanassiou M, Theodossiadis PG, Liarakos VS, Rouvas A, Giamarellos-Bourboulis EJ, Vergados IA. Inhibition of corneal neovascularization by subconjunctival bevacizumab in an animal model. Am J Ophthalmol. 2008;145:424–31.
    1. Eskens FA, Verweij J. The clinical toxicity profile of vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor (VEGFR) targeting angiogenesis inhibitors; a review. Eur J Cancer. 2006;42:3127–39.
    1. Bock F, Onderka J, Rummelt C, Dietrich T, Bachmann B, Kruse FE, Schlotzer-Schrehardt U, Cursiefen C. Safety profile of topical VEGF neutralization at the cornea. Invest Ophthalmol Vis Sci. 2009;50:2095–102.
    1. Ferrara N. The role of VEGF in the regulation of physiological and pathological angiogenesis. EXS. 2005;94:209–31.
    1. Ferrara N. Vascular endothelial growth factor: basic science and clinical progress. Endocr Rev. 2004;25:581–611.
    1. Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov. 2004;3:391–400.
    1. Presta LG, Chen H, O'Connor SJ, Chisholm V, Meng YG, Krummen L, Winkler M, Ferrara N. Humanization of an anti-vascular endothelial growth factor monoclonal antibody for the therapy of solid tumors and other disorders. Cancer Res. 1997;57:4593–9.
    1. Barkmeier AJ, Akduman L. Bevacizumab (avastin) in ocular processes other than choroidal neovascularization. Ocul Immunol Inflamm. 2009;17:109–17.
    1. Grisanti S, Ziemssen F. Bevacizumab: off-label use in ophthalmology. Indian J Ophthalmol. 2007;55:417–20.
    1. Wickremasinghe SS, Michalova K, Gilhotra J, Guymer RH, Harper CA, Wong TY, Qureshi S. Acute intraocular inflammation after intravitreous injections of bevacizumab for treatment of neovascular age-related macular degeneration. Ophthalmology. 2008;115:1911–5.
    1. DeStafeno JJ, Kim T. Topical bevacizumab therapy for corneal neovascularization. Arch Ophthalmol. 2007;125:834–6.
    1. Kim SW, Ha BJ, Kim EK, Tchah H, Kim TI. The effect of topical bevacizumab on corneal neovascularization. Ophthalmology. 2008;115:e33–8.
    1. Uy HS, Chan PS, Ang RE. Topical bevacizumab and ocular surface neovascularization in patients with stevens-johnson syndrome. Cornea. 2008;27:70–3.
    1. Erdurmus M, Totan Y. Subconjunctival bevacizumab for corneal neovascularization. Graefes Arch Clin Exp Ophthalmol. 2007;245:1577–9.
    1. Doctor PP, Bhat PV, Foster CS. Subconjunctival bevacizumab for corneal neovascularization. Cornea. 2008;27:992–5.
    1. Bahar I, Kaiserman I, McAllum P, Rootman D, Slomovic A. Subconjunctival bevacizumab injection for corneal neovascularization in recurrent pterygium. Curr Eye Res. 2008;33:23–8.
    1. Dastjerdi MH, Al-Arfaj KM, Nallasamy N, Hamrah P, Jurkunas UV, Pineda R, 2nd, Pavan-Langston D, Dana R. Topical bevacizumab in the treatment of corneal neovascularization: results of a prospective, open-label, noncomparative study. Arch Ophthalmol. 2009;127:381–9.
    1. Jacobs DS, Lim M, Carrasquillo KG, Rosenthal P. Bevacizumab for corneal neovascularization. Ophthalmology. 2009;116:592–3. author reply 3-4.
    1. Mackenzie SE, Tucker WR, Poole TR. Bevacizumab (Avastin) for corneal neovascularization--corneal light shield soaked application. Cornea. 2009;28:246–7.
    1. Oh JY, Kim MK, Shin MS, Lee HJ, Lee JH, Wee WR. The anti-inflammatory effect of subconjunctival bevacizumab on chemically burned rat corneas. Curr Eye Res. 2009;34:85–91.
    1. You IC, Kang IS, Lee SH, Yoon KC. Therapeutic effect of subconjunctival injection of bevacizumab in the treatment of corneal neovascularization. Acta Ophthalmol. 2008
    1. Han YS, Lee JE, Jung JW, Lee JS. Inhibitory effects of bevacizumab on angiogenesis and corneal neovascularization. Graefes Arch Clin Exp Ophthalmol. 2009;247:541–8.
    1. Chen WL, Lin CT, Lin NT, Tu IH, Li JW, Chow LP, Liu KR, Hu FR. Subconjunctival injection of bevacizumab (avastin) on corneal neovascularization in different rabbit models of corneal angiogenesis. Invest Ophthalmol Vis Sci. 2009;50:1659–65.
    1. Duch S, Buchacra O, Milla E, Milla E, Andreu D, Tellez J. Intracameral bevacizumab (Avastin) for neovascular glaucoma: a pilot study in 6 patients. J Glaucoma. 2009;18:140–3.
    1. Costagliola C, Cipollone U, Rinaldi M, della Corte M, Semeraro F, Romano MR. Intravitreal bevacizumab (Avastin) injection for neovascular glaucoma: a survey on 23 cases throughout 12-month follow-up. Br J Clin Pharmacol. 2008;66:667–73.
    1. Kovacevic D, Caljkusic-Mance T, Misljenovic T, Mikulicic M, Alpeza-Dunato Z. Intravitreal bevacizumab for the management of age-related macular degeneration. Coll Antropol. 2008;32(Suppl 2):5–7.
    1. Elayappan B, Ravinarayannan H, Sardar Pasha SP, Lee KJ, Gurunathan S. PEDF inhibits VEGF- and EPO- induced angiogenesis in retinal endothelial cells through interruption of PI3K/Akt phosphorylation. Angiogenesis 2009.
    1. Chan WM, Lai TY, Chan KP, Li H, Liu DT, Lam DS, Pang CP. Changes in aqueous vascular endothelial growth factor and pigment epithelial-derived factor levels following intravitreal bevacizumab injections for choroidal neovascularization secondary to age-related macular degeneration or pathologic myopia. Retina. 2008;28:1308–13.
    1. van Wijngaarden P, Brereton HM, Gibbins IL, Coster DJ, Williams KA. Kinetics of strain-dependent differential gene expression in oxygen-induced retinopathy in the rat. Exp Eye Res. 2007;85:508–17.
    1. Browning AC, Dua HS, Amoaku WM. The effects of growth factors on the proliferation and in vitro angiogenesis of human macular inner choroidal endothelial cells. Br J Ophthalmol. 2008;92:1003–8.
    1. Economou MA, Wu J, Vasilcanu D, Rosengren L, All-Ericsson C, van der Ploeg I, Menu E, Girnita L, Axelson M, Larsson O, Seregard S, Kvanta A. Inhibition of VEGF secretion and experimental choroidal neovascularization by picropodophyllin (PPP), an inhibitor of the insulin-like growth factor-1 receptor. Invest Ophthalmol Vis Sci. 2008;49:2620–6.
    1. Moldobaeva A, Baek A, Wagner EM. MIP-2 causes differential activation of RhoA in mouse aortic versus pulmonary artery endothelial cells. Microvasc Res. 2008;75:53–8.
    1. Kollmar O, Scheuer C, Menger MD, Schilling MK. Macrophage inflammatory protein-2 promotes angiogenesis, cell migration, and tumor growth in hepatic metastasis. Ann Surg Oncol. 2006;13:263–75.
    1. Xue ML, Thakur A, Willcox M. Macrophage inflammatory protein-2 and vascular endothelial growth factor regulate corneal neovascularization induced by infection with Pseudomonas aeruginosa in mice. Immunol Cell Biol. 2002;80:323–7.
    1. Lu P, Li L, Mukaida N, Zhang X. Alkali-induced corneal neovascularization is independent of CXCR2-mediated neutrophil infiltration. Cornea. 2007;26:199–206.
    1. Yu L, Wu X, Cheng Z, Lee CV, LeCouter J, Campa C, Fuh G, Lowman H, Ferrara N. Interaction between bevacizumab and murine VEGF-A: a reassessment. Invest Ophthalmol Vis Sci. 2008;49:522–7.
    1. Mahoney JM, Waterbury LD. Drug effects on the neovascularization response to silver nitrate cauterization of the rat cornea. Curr Eye Res. 1985;4:531–5.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)). Methods. 2001;25:402–8.
    1. Regenfuss B, Bock F, Parthasarathy A, Cursiefen C. Corneal (lymph)angiogenesis--from bedside to bench and back: a tribute to Judah Folkman. Lymphat Res Biol. 2008;6:191–201.
    1. Peyman GA, Kivilcim M, Morales AM, DellaCroce JT, Conway MD. Inhibition of corneal angiogenesis by ascorbic acid in the rat model. Graefes Arch Clin Exp Ophthalmol. 2007;245:1461–7.
    1. Hurmeric V, Mumcuoglu T, Erdurman C, Kurt B, Dagli O, Durukan AH. Effect of subconjunctival bevacizumab (Avastin) on experimental corneal neovascularization in guinea pigs. Cornea. 2008;27:357–62.
    1. Naoumidi TL, Pallikaris IG, Naoumidi II, Astyrakakis NI. Conductive keratoplasty: histological study of human corneas. Am J Ophthalmol. 2005;140:984–92.
    1. Barros LF, Belfort R., Jr The effects of the subconjunctival injection of bevacizumab (Avastin) on angiogenesis in the rat cornea. An Acad Bras Cienc. 2007;79:389–94.
    1. Kim TI, Kim SW, Kim S, Kim T, Kim EK. Inhibition of experimental corneal neovascularization by using subconjunctival injection of bevacizumab (Avastin). Cornea. 2008;27:349–52.
    1. Cakir M, Cekic O, Yilmaz OF. Intravitreal bevacizumab and triamcinolone treatment for choroidal neovascularization in Best disease. J AAPOS. 2009;13:94–6.
    1. Beutel J, Ziemssen F, Luke M, Partsch M, Bartz-Schmidt KU, Gelisken F. Intravitreal bevacizumab treatment of macular edema in central retinal vein occlusion: one-year results. Int Ophthalmol 2008.
    1. Bakri SJ, Snyder MR, Reid JM, Pulido JS, Ezzat MK, Singh RJ. Pharmacokinetics of intravitreal ranibizumab (Lucentis). Ophthalmology. 2007;114:2179–82.
    1. Dib E, Maia M, Longo-Maugeri IM, Martins MC, Mussalem JS, Squaiella CC, Penha FM, Magalhaes O, Jr, Rodrigues EB, Farah ME. Subretinal bevacizumab detection after intravitreous injection in rabbits. Invest Ophthalmol Vis Sci. 2008;49:1097–100.
    1. Montezuma SR, Vavvas D, Miller JW. Review of the ocular angiogenesis animal models. Semin Ophthalmol. 2009;24:52–61.
    1. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003;9:669–76.
    1. Yan J, Zeng Y, Jiang J, Zhou J, Yin Z, Wang Z, Zhu P. The expression patterns of vascular endothelial growth factor and thrombospondin 2 after corneal alkali burn. Colloids Surf B Biointerfaces. 2007;60:105–9.
    1. Tombran-Tink J, Aparicio S, Xu X, Tink AR, Lara N, Sawant S, Barnstable CJ, Zhang SS. PEDF and the serpins: phylogeny, sequence conservation, and functional domains. J Struct Biol. 2005;151:130–50.
    1. Tombran-Tink J, Barnstable CJ. PEDF: a multifaceted neurotrophic factor. Nat Rev Neurosci. 2003;4:628–36.
    1. Tombran-Tink J, Barnstable CJ. Therapeutic prospects for PEDF: more than a promising angiogenesis inhibitor. Trends Mol Med. 2003;9:244–50.
    1. Harper SJ, Bates DO. VEGF-A splicing: the key to anti-angiogenic therapeutics? Nat Rev Cancer. 2008;8:880–7.
    1. Sivakumar V, Zhang Y, Ling EA, Foulds WS, Kaur C. Insulin-like growth factors, angiopoietin-2, and pigment epithelium-derived growth factor in the hypoxic retina. J Neurosci Res. 2008;86:702–11.
    1. Kvanta A. Neovascular age-related macular degeneration: too many theories, too little knowledge? Acta Ophthalmol. 2008;86:468–9.
    1. Norrby K. Interleukin-8 and de novo mammalian angiogenesis. Cell Prolif. 1996;29:315–23.
    1. Norrby K. Vascular endothelial growth factor and de novo mammalian angiogenesis. Microvasc Res. 1996;51:153–63.

Source: PubMed

3
Tilaa