Topically Applied Etamsylate: A New Orphan Drug for HHT-Derived Epistaxis (Antiangiogenesis through FGF Pathway Inhibition)

Virginia Albiñana, Guillermo Giménez-Gallego, Angela García-Mato, Patricia Palacios, Lucia Recio-Poveda, Angel-M Cuesta, José-Luis Patier, Luisa-María Botella, Virginia Albiñana, Guillermo Giménez-Gallego, Angela García-Mato, Patricia Palacios, Lucia Recio-Poveda, Angel-M Cuesta, José-Luis Patier, Luisa-María Botella

Abstract

Hereditary hemorrhagic telangiectasia (HHT) is a vascular dysplasia characterized by recurrent and spontaneous epistaxis (nose bleeds), telangiectases on skin and mucosa, internal organ arteriovenous malformations, and dominant autosomal inheritance. Mutations in Endoglin and ACVRL1 / ALK1 , genes mainly expressed in endothelium, are responsible in 90% of the cases for the pathology. These genes are involved in the transforming growth factor-β(TGF-β) signaling pathway. Epistaxis remains as one of the most common symptoms impairing the quality of life of patients, becoming life-threatening in some cases. Different strategies have been used to decrease nose bleeds, among them is antiangiogenesis. The two main angiogenic pathways in endothelial cells depend on vascular endothelial growth factor and fibroblast growth factor (FGF). The present work has used etamsylate, the diethylamine salt of the 2,5-dihydroxybenzene sulfonate anion, also known as dobesilate, as a FGF signaling inhibitor. In endothelial cells, in vitro experiments show that etamsylate acts as an antiangiogenic factor, inhibiting wound healing and matrigel tubulogenesis. Moreover, etamsylate decreases phosphorylation of Akt and ERK1/2. A pilot clinical trial (EudraCT: 2016-003982-24) was performed with 12 HHT patients using a topical spray of etamsylate twice a day for 4 weeks. The epistaxis severity score (HHT-ESS) and other pertinent parameters were registered in the clinical trial. The significant reduction in the ESS scale, together with the lack of significant side effects, allowed the designation of topical etamsylate as a new orphan drug for epistaxis in HHT (EMA/OD/135/18).

Keywords: Alk1; Endoglin; FGF pathway; HHT; TGF-β signaling pathway; antiangiogenesis; epistaxis; etamsylate.

Conflict of interest statement

Conflict of Interest None declared.

Figures

Fig. 1
Fig. 1
Effect of etamsylate on angiogenesis in HMEC-1 cells. (A) Wound healing assay. In the upper part, representative images of the different experiments and replicas are shown. In the lower part, the percentage of migration in time is shown at different doses. (B) Tubulogenesis assay. In the upper part, representative images are shown. In the lower part, the tube densities per field at different doses compared with the untreated cells are shown. Mean with the standard deviation is represented normalized versus control. Statistical significance was studied using Student'st-test. No significant differences were found in this case (ns).
Fig. 2
Fig. 2
Effect of etamsylate on angiogenesis in HUVECs. (A) Wound-healing assay. In the representative images shown, the cell migration after etamsylate administration is observed. In the lower part, the percentage of wound healing (migration) in time is represented. The mean with SD is shown. Values are normalized with control. Statistical significance was studied using Student'st-test. *p < 0.05. (B) Tubulogenesis assay. Representative images are shown, demonstrating a lower amount of tubes in the treated cells compared with untreated cells. The different tube densities are shown in a representative experiment. The mean with SD is shown. Values are normalized with control. Statistical significance was studied using Student'st-test. *p < 0.5. HUVECs, human umbilical vein endothelial cells.
Fig. 3
Fig. 3
(A) Effect of etamsylate on tubulogenesis assay in BOECs. Representative images demonstrate a lower amount of tubes in the treated cells. The different tube densities are shown in the graph. The number of closed tubules was counted in five different fields and the mean was calculated. The mean with SD is shown. Statistical significance was studied using Student'st-test (healthy:p < 0.002; HHT:p < 0.023). (B) Tubulogenesis assay FGF vs. FGF/etamsylate. HHT BOECs were treated with or without FGF and etamsylate. The number of closed tubules was counted in five different fields and the mean was calculated. The images and graph show a significant decrease in the number of closed tubes when cells are without FGF (***p < 2.6 × 10 −6 ), and when etamsylate is added in the presence of FGF (FGF/etamsylate; ***p < 1.1 × 10 −6 ) in comparison with FGF-treated cells (FGF). BOECs, blood outgrowth endothelial cells; FGF, fibroblast growth factor; HHT, hereditary hemorrhagic telangiectasia.
Fig. 4
Fig. 4
3D angiogenesis. HUVEC-coated beads were cultured in the presence or absence of etamsylate during 5 days. When etamsylate is present, fewer bonds can be observed between neighboring beads forming a network, as it occurs in control conditions. There was a significantly higher number of ramifications in control (untreated cells) compared with etamsylate-treated cells, as the graph shows (day 4, ***p < 6.5 × 10 −4 ; day 5, ***p = 8.34 × 10 −5 ). HUVECs, human umbilical vein endothelial cells.
Fig. 5
Fig. 5
Effect of etamsylate on viability and Endoglin and ALK1 RNA expression. (A) Viability of HUVECs and BOECs cells in the 0 to 100 μM range of etamsylate treatment is not affected. (B) RNA expression of ENG and ALK1 after treatment with different doses of etamsylate in HUVECs. RNA levels ofACVRL1/ALK1andEndoglinwere determined by RT-qPCR in HUVECs, after etamsylate treatments (range: 10–100 μM), and normalized with the untreated controls. Statistical significance was studied using Student'st-test. No significant difference was found. BOECs, blood outgrowth endothelial cells; HUVECs, human umbilical vein endothelial cells.
Fig. 6
Fig. 6
Analysis of Akt and Erk1 phosphorylation after etamsylate treatment. (A) Analysis of pAkt1, Akt1, and tubulin in cell lysates in HUVECs. A representative gel of three experiments performed is shown. Bands were quantified by Adobe Photoshop CS. Relative levels of pAkt1 normalized versus total Akt1. A decrease of the amount of phosphorylated protein is apparent compared with control. (B) Relative levels of pERK2 normalized versus total erk. pERK2 levels decrease when cells were treated with the highest doses of etamsylate (50–100 μM) compared with the control. HUVECs, human umbilical vein endothelial cells.
Fig. 7
Fig. 7
Levels of epistaxis severity score (ESS). (A) HHT-ESS of the 12 patients before and after the treatment. (B) HHT-ESS mean of pre- and posttreatment levels of the patients. A Student'st-test was made to compare pre- and posttreatment HHT-ESS values. Differences were significant atp < 0.01. (CandD) Analysis of FGF and VEGF, as biomarkers related to angiogenesis and bleeding. The graph shows pre- and posttreatment results. FGF, fibroblast growth factor; HHT, hereditary hemorrhagic telangiectasia; VEGF, vascular endothelial growth factor.
Fig. 8
Fig. 8
The antiangiogenic effect of etamsylate on the nose mucosa. Aspect of the nose mucosa in a patient before starting the treatment, and around the same area after 4 weeks of treatment with etamsylate (* shows the major affected area).

References

    1. Abdalla S A, Letarte M. Hereditary haemorrhagic telangiectasia: current views on genetics and mechanisms of disease. J Med Genet. 2006;43(02):97–110.
    1. Govani F S, Shovlin C L. Hereditary haemorrhagic telangiectasia: a clinical and scientific review. Eur J Hum Genet. 2009;17(07):860–871.
    1. Shovlin C L. Hereditary haemorrhagic telangiectasia: pathophysiology, diagnosis and treatment. Blood Rev. 2010;24(06):203–219.
    1. Shovlin C L, Buscarini E, Kjeldsen A D et al.European Reference Network for Rare Vascular Diseases (VASCERN) outcome measures for hereditary haemorrhagic telangiectasia (HHT) Orphanet J Rare Dis. 2018;13(01):136–140.
    1. Shovlin C L, Guttmacher A E, Buscarini E et al.Diagnostic criteria for hereditary hemorrhagic telangiectasia (Rendu-Osler-Weber syndrome) Am J Med Genet. 2000;91(01):66–67.
    1. Plauchu H, de Chadarévian J P, Bideau A, Robert J M. Age-related clinical profile of hereditary hemorrhagic telangiectasia in an epidemiologically recruited population. Am J Med Genet. 1989;32(03):291–297.
    1. McAllister K A, Grogg K M, Johnson D W et al.Endoglin, a TGF-beta binding protein of endothelial cells, is the gene for hereditary haemorrhagic telangiectasia type 1. Nat Genet. 1994;8(04):345–351.
    1. Johnson D W, Berg J N, Baldwin M A et al.Mutations in the activin receptor-like kinase 1 gene in hereditary haemorrhagic telangiectasia type 2. Nat Genet. 1996;13(02):189–195.
    1. Cole S G, Begbie M E, Wallace G M, Shovlin C L. A new locus for hereditary haemorrhagic telangiectasia (HHT3) maps to chromosome 5. J Med Genet. 2005;42(07):577–582.
    1. Bayrak-Toydemir P, McDonald J, Akarsu N et al.A fourth locus for hereditary hemorrhagic telangiectasia maps to chromosome 7. Am J Med Genet A. 2006;140(20):2155–2162.
    1. Wooderchak-Donahue W L, McDonald J, O'Fallon B et al.BMP9 mutations cause a vascular-anomaly syndrome with phenotypic overlap with hereditary hemorrhagic telangiectasia. Am J Hum Genet. 2013;93(03):530–537.
    1. Albiñana V, Recio-Poveda L, Zarrabeitia R, Botella L M. Current and emerging pharmacotherapies for hereditary hemorrhagic telangiectasia. Expert Opin Orphan Drugs. 2017;5:665–675.
    1. Albiñana V, Bernabeu-Herrero M E, Zarrabeitia R, Bernabéu C, Botella L M. Estrogen therapy for hereditary haemorrhagic telangiectasia (HHT): effects of raloxifene, on Endoglin and ALK1 expression in endothelial cells. Thromb Haemost. 2010;103(03):525–534.
    1. Albiñana V, Sanz-Rodríguez F, Recio-Poveda L, Bernabéu C, Botella L M. Immunosuppressor FK506 increases endoglin and activin receptor-like kinase 1 expression and modulates transforming growth factor-β1 signaling in endothelial cells. Mol Pharmacol. 2011;79(05):833–843.
    1. Zarrabeitia R, Ojeda-Fernandez L, Recio L et al.Bazedoxifene, a new orphan drug for the treatment of bleeding in hereditary haemorrhagic telangiectasia. Thromb Haemost. 2016;115(06):1167–1177.
    1. Albiñana V, Recio-Poveda L, Zarrabeitia R, Bernabéu C, Botella L M. Propranolol as antiangiogenic candidate for the therapy of hereditary haemorrhagic telangiectasia. Thromb Haemost. 2012;108(01):41–53.
    1. Fernández I S, Cuevas P, Angulo J et al.Gentisic acid, a compound associated with plant defense and a metabolite of aspirin, heads a new class of in vivo fibroblast growth factor inhibitors. J Biol Chem. 2010;285(15):11714–11729.
    1. Olsen S K, Garbi M, Zampieri N et al.Fibroblast growth factor (FGF) homologous factors share structural but not functional homology with FGFs. J Biol Chem. 2003;278(36):34226–34236.
    1. Itoh N, Ornitz D M. Evolution of the Fgf and Fgfr gene families. Trends Genet. 2004;20(11):563–569.
    1. Baird A, Bohlen P.Peptide growth factors and their receptors Berlin: Springer-Verlag; 1990. 95:369–418.
    1. Giménez-Gallego G, Cuevas P. Fibroblast growth factors, proteins with a broad spectrum of biological activities. Neurol Res. 1994;16(04):313–316.
    1. Cuevas P, García-Calvo M, Carceller F et al.Correction of hypertension by normalization of endothelial levels of fibroblast growth factor and nitric oxide synthase in spontaneously hypertensive rats. Proc Natl Acad Sci U S A. 1996;93(21):11996–12001.
    1. Cuevas P, Reimers D, Carceller F, Xiaobing F, Gimenez-Gallego G. New York, NY: Plenum Press; 1996. Second messengers and protein phosphorylation in health and disease; pp. 161–167.
    1. Abuharbeid S, Czubayko F, Aigner A. The fibroblast growth factor-binding protein FGF-BP. Int J Biochem Cell Biol. 2006;38(09):1463–1468.
    1. Finetti F, Solito R, Morbidelli L, Giachetti A, Ziche M, Donnini S. Prostaglandin E2 regulates angiogenesis via activation of fibroblast growth factor receptor-1. J Biol Chem. 2008;283(04):2139–2146.
    1. Jackson A, Friedman S, Zhan X, Engleka K A, Forough R, Maciag T. Heat shock induces the release of fibroblast growth factor 1 from NIH 3T3 cells. Proc Natl Acad Sci U S A. 1992;89(22):10691–10695.
    1. Shin J T, Opalenik S R, Wehby J N et al.Serum-starvation induces the extracellular appearance of FGF-1. Biochim Biophys Acta. 1996;1312(01):27–38.
    1. Matsunaga H, Ueda H. Voltage-dependent N-type Ca2+ channel activity regulates the interaction between FGF-1 and S100A13 for stress-induced non-vesicular release. Cell Mol Neurobiol. 2006;26(03):237–246.
    1. Thomas K A, Rios-Candelore M, Giménez-Gallego G et al.Pure brain-derived acidic fibroblast growth factor is a potent angiogenic vascular endothelial cell mitogen with sequence homology to interleukin 1. Proc Natl Acad Sci U S A. 1985;82(19):6409–6413.
    1. Esch F, Baird A, Ling N et al.Primary structure of bovine pituitary basic fibroblast growth factor (FGF) and comparison with the amino-terminal sequence of bovine brain acidic FGF. Proc Natl Acad Sci U S A. 1985;82(19):6507–6511.
    1. Hanahan D, Weinberg R A. The hallmarks of cancer. Cell. 2000;100(01):57–70.
    1. Jonca F, Ortéga N, Gleizes P E, Bertrand N, Plouët J. Cell release of bioactive fibroblast growth factor 2 by exon 6-encoded sequence of vascular endothelial growth factor. J Biol Chem. 1997;272(39):24203–24209.
    1. Pàez-Ribes M, Allen E, Hudock J et al.Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. 2009;15(03):220–231.
    1. Loges S, Mazzone M, Hohensinner P, Carmeliet P. Silencing or fueling metastasis with VEGF inhibitors: antiangiogenesis revisited. Cancer Cell. 2009;15(03):167–170.
    1. Fernández-Tornero C, Lozano R M, Redondo-Horcajo M et al.Leads for development of new naphthalenesulfonate derivatives with enhanced antiangiogenic activity: crystal structure of acidic fibroblast growth factor in complex with 5-amino-2-naphthalene sulfonate. J Biol Chem. 2003;278(24):21774–21781.
    1. Andrés G, Leali D, Mitola Set al.A pro-inflammatory signature mediates FGF2-induced angiogenesis J Cell Mol Med 200913(8B):2083–2108.
    1. Cha Y I, DuBois R N. NSAIDs and cancer prevention: targets downstream of COX-2. Annu Rev Med. 2007;58:239–252.
    1. Haritoglou C, Gerss J, Sauerland C, Kampik A, Ulbig M W; CALDIRET study group.Effect of calcium dobesilate on occurrence of diabetic macular oedema (CALDIRET study): randomised, double-blind, placebo-controlled, multicentre trial Lancet 2009373(9672):1364–1371.
    1. Zhang X, Liu W, Wu S, Jin J, Li W, Wang N. Calcium dobesilate for diabetic retinopathy: a systematic review and meta-analysis. Sci China Life Sci. 2015;58(01):101–107.
    1. Angulo J, Peiró C, Romacho Tet al.Inhibition of vascular endothelial growth factor (VEGF)-induced endothelial proliferation, arterial relaxation, vascular permeability and angiogenesis by dobesilate Eur J Pharmacol 2011667(1–3):153–159.
    1. Cuevas P, Díaz-González D, García-Martín-Córdova C et al.Dobesilate diminishes activation of the mitogen-activated protein kinase ERK1/2 in glioma cells. J Cell Mol Med. 2006;10(01):225–230.
    1. Cuevas P, Carceller F, Angulo J, González-Corrochano R, Cuevas-Bourdier A, Giménez-Gallego G. Antiglioma effects of a new, low molecular mass, inhibitor of fibroblast growth factor. Neurosci Lett. 2011;491(01):1–7.
    1. Fernandez-L A, Sanz-Rodriguez F, Zarrabeitia R et al.Blood outgrowth endothelial cells from hereditary haemorrhagic Telangiectasia patients reveal abnormalities compatible with vascular lesions. Cardiovasc Res. 2005;68(02):235–248.
    1. Fernandez-L A, Garrido-Martin E M, Sanz-Rodriguez F et al.Gene expression fingerprinting for human hereditary hemorrhagic telangiectasia. Hum Mol Genet. 2007;16(13):1515–1533.
    1. Hoag J B, Terry P, Mitchell S, Reh D, Merlo C A. An epistaxis severity score for hereditary hemorrhagic telangiectasia. Laryngoscope. 2010;120(04):838–843.
    1. Somanath P R, Razorenova O V, Chen J, Byzova T V. Akt1 in endothelial cell and angiogenesis. Cell Cycle. 2006;5(05):512–518.
    1. Hata Y, Rook S L, Aiello L P. Basic fibroblast growth factor induces expression of VEGF receptor KDR through a protein kinase C and p44/p42 mitogen-activated protein kinase-dependent pathway. Diabetes. 1999;48(05):1145–1155.
    1. Murakami M, Nguyen L T, Hatanaka K et al.FGF-dependent regulation of VEGF receptor 2 expression in mice. J Clin Invest. 2011;121(07):2668–2678.
    1. Shovlin C L, Sulaiman N L, Govani F S, Jackson J E, Begbie M E. Elevated factor VIII in hereditary haemorrhagic telangiectasia (HHT): association with venous thromboembolism. Thromb Haemost. 2007;98(05):1031–1039.

Source: PubMed

3
Iratkozz fel