Pazopanib may reduce bleeding in hereditary hemorrhagic telangiectasia

Marie E Faughnan, James R Gossage, Murali M Chakinala, S Paul Oh, Raj Kasthuri, Christopher C W Hughes, Justin P McWilliams, Joseph G Parambil, Nicholas Vozoris, Jill Donaldson, Gitanjali Paul, Pamela Berry, Dennis L Sprecher, Marie E Faughnan, James R Gossage, Murali M Chakinala, S Paul Oh, Raj Kasthuri, Christopher C W Hughes, Justin P McWilliams, Joseph G Parambil, Nicholas Vozoris, Jill Donaldson, Gitanjali Paul, Pamela Berry, Dennis L Sprecher

Abstract

Pazopanib (Votrient) is an orally administered tyrosine kinase inhibitor that blocks VEGF receptors potentially serving as anti-angiogenic treatment for hereditary hemorrhagic telangiectasia (HHT). We report a prospective, multi-center, open-label, dose-escalating study [50 mg, 100 mg, 200 mg, and 400 mg], designed as a proof-of-concept study to demonstrate efficacy of pazopanib on HHT-related bleeding, and to measure safety. Patients, recruited at 5 HHT Centers, required ≥ 2 Curacao criteria AND [anemia OR severe epistaxis with iron deficiency]. Co-primary outcomes, hemoglobin (Hgb) and epistaxis severity, were measured during and after treatment, and compared to baseline. Safety monitoring occurred every 1.5 weeks. Seven patients were treated with 50 mg pazopanib daily. Six/seven showed at least 50% decrease in epistaxis duration relative to baseline at some point during study; 3 showed at least 50% decrease in duration during Weeks 11 and 12. Six patients showed a decrease in ESS of > 0.71 (MID) relative to baseline at some point during study; 3/6 showed a sustained improvement. Four patients showed > 2 gm improvement in Hgb relative to baseline at one or more points during study. Health-related QOL scores improved on all SF-36 domains at Week 6 and/or Week 12, except general health (unchanged). There were 19 adverse events (AE) including one severe AE (elevated LFTs, withdrawn from dosing at 43 days); with no serious AE. In conclusion, we observed an improvement in Hgb and/or epistaxis in all treated patients. This occurred at a dose much lower than typically used for oncologic indications, with no serious AE. Further studies of pazopanib efficacy are warranted.

Keywords: Anemia; Epistaxis; Hereditary; Telangiectasia; Tyrosine kinase inhibitor.

Conflict of interest statement

MF reports grants from GSK, during the conduct of the study; personal fees from HHT Foundation International (Cure HHT), grants from NIH-NINDS-NCATS, grants from NIH-NHLBI, grants from US Department of Defense, outside the submitted work; MCM reports grants from GSK, during the conduct of the study; grants from HHT Foundation International, personal fees from Express Scripts International, outside the submitted work. JD is a GSK employee and share holder. GP reports other from Novartis, during the conduct of the study; other from GSK, outside the submitted work; PB reports other from GSK, other from Janssen Global Services, outside the submitted work. DLS reports other from GlaxoSmithKline, outside the submitted work; and I have worked with the HHT patient advocacy group for many years, but receive no monies related to that activity. However, this has motivated my interest in pursuing this work; JG, SPO, RK, CCWH, JM, JGP, NV have nothing to disclose.

Figures

Fig. 1
Fig. 1
HB (gm/dL) and duration of epistaxis (min/2 weeks) for each patient, Reported at baseline, during therapy, and post-treatment follow-up
Fig. 1
Fig. 1
HB (gm/dL) and duration of epistaxis (min/2 weeks) for each patient, Reported at baseline, during therapy, and post-treatment follow-up
Fig. 2
Fig. 2
Epistaxis severity score (ESS) for each patient, with group means, reported at baseline, after 6 and 12 weeks of therapy, and at follow-up. The dashed line represents the time period after drug discontinuation in Patient 5

References

    1. Donaldson JW, McKeever TM, Hall IP, Hubbard RB, Fogarty AW. The UK prevalence of hereditary haemorrhagic telangiectasia and its association with sex, socioeconomic status and region of residence: a population-based study. Thorax. 2014;69(2):161–167. doi: 10.1136/thoraxjnl-2013-203720.
    1. McAllister KA, Grogg KM, Johnson DW, Gallione CJ, Baldwin MA, Jackson CE, et al. Endoglin, a TGF-beta binding protein of endothelial cells, is the gene for hereditary haemorrhagic telangiectasia type 1. Nat Genet. 1994;8(4):345–351. doi: 10.1038/ng1294-345.
    1. Johnson DW, Berg JN, Baldwin MA, Gallione CJ, Marondel I, Yoon SJ, et al. Mutations in the activin receptor-like kinase 1 gene in hereditary haemorrhagic telangiectasia type 2. Nat Genet. 1996;13(2):189–195. doi: 10.1038/ng0696-189.
    1. Gallione CJ, Repetto GM, Legius E, Rustgi AK, Schelley SL, Tejpar S, et al. A combined syndrome of juvenile polyposis and hereditary haemorrhagic telangiectasia associated with mutations in MADH4 (SMAD4) Lancet. 2004;363(9412):852–859. doi: 10.1016/S0140-6736(04)15732-2.
    1. Ingrosso M, Sabba C, Pisani A, Principi M, Gallitelli M, Cirulli A, et al. Evidence of small-bowel involvement in hereditary hemorrhagic telangiectasia: a capsule-endoscopic study. Endoscopy. 2004;36(12):1074–1079. doi: 10.1055/s-2004-826045.
    1. Geisthoff UW, Heckmann K, D’Amelio R, Grunewald S, Knobber D, Falkai P, et al. Health-related quality of life in hereditary hemorrhagic telangiectasia. Otolaryngol Head Neck Surg. 2007;136(5):726–733. doi: 10.1016/j.otohns.2006.12.019.
    1. Merlo CA, Yin LX, Hoag JB, Mitchell SE, Reh DD. The effects of epistaxis on health-related quality of life in patients with hereditary hemorrhagic telangiectasia. Int Forum Allergy Rhinol. 2014;4(11):921–925. doi: 10.1002/alr.21374.
    1. Brinjikji W, Wood CP, Lanzino G, Cloft HJ, Misra S, Kallmes DF, et al. High rates of bleeding complications among hospitalized patients with hereditary hemorrhagic telangiectasia in the United States. Ann Am Thorac Soc. 2016;13(9):1505–1511. doi: 10.1513/AnnalsATS.201603-200OC.
    1. Kasthuri RS, Montifar M, Nelson J, Kim H, Lawton MT, Faughnan ME, et al. Prevalence and predictors of anemia in hereditary hemorrhagic telangiectasia. Am J Hematol. 2017;92:E591–E593. doi: 10.1002/ajh.24832.
    1. Whitehead KJ, Sautter NB, McWilliams JP, Chakinala MM, Merlo CA, Johnson MH, et al. Effect of topical intranasal therapy on epistaxis frequency in patients with hereditary hemorrhagic telangiectasia: a randomized clinical trial. JAMA. 2016;316(9):943–951. doi: 10.1001/jama.2016.11724.
    1. Geisthoff UW, Seyfert UT, Kubler M, Bieg B, Plinkert PK, Konig J. Treatment of epistaxis in hereditary hemorrhagic telangiectasia with tranexamic acid—a double-blind placebo-controlled cross-over phase IIIB study. Thromb Res. 2014;134(3):565–571. doi: 10.1016/j.thromres.2014.06.012.
    1. Yaniv E, Preis M, Shevro J, Nageris B, Hadar T. Anti-estrogen therapy for hereditary hemorrhagic telangiectasia—a long-term clinical trial. Rhinology. 2011;49(2):214–216.
    1. Gaillard S, Dupuis-Girod S, Boutitie F, Riviere S, Moriniere S, Hatron PY, et al. Tranexamic acid for epistaxis in hereditary hemorrhagic telangiectasia patients: a European cross-over controlled trial in a rare disease. J Thromb Haemost. 2014;12(9):1494–1502. doi: 10.1111/jth.12654.
    1. Karapantzos I, Tsimpiris N, Goulis DG, Van Hoecke H, Van Cauwenberge P, Danielides V. Management of epistaxis in hereditary hemorrhagic telangiectasia by Nd:YAG laser and quality of life assessment using the HR-QoL questionnaire. Eur Arch Otorhinolaryngol. 2005;262(10):830–833. doi: 10.1007/s00405-004-0911-0.
    1. Hanks JE, Hunter D, Goding GS, Jr, Boyer HC. Complications from office sclerotherapy for epistaxis due to hereditary hemorrhagic telangiectasia (HHT or Osler-Weber-Rendu) Int Forum Allergy Rhinol. 2014;4(5):422–427. doi: 10.1002/alr.21287.
    1. Longacre AV, Gross CP, Gallitelli M, Henderson KJ, White RI, Jr, Proctor DD. Diagnosis and management of gastrointestinal bleeding in patients with hereditary hemorrhagic telangiectasia. Am J Gastroenterol. 2003;98(1):59–65. doi: 10.1111/j.1572-0241.2003.07185.x.
    1. Sadick H, Naim R, Sadick M, Hormann K, Riedel F. Plasma level and tissue expression of angiogenic factors in patients with hereditary hemorrhagic telangiectasia. Int J Mol Med. 2005;15(4):591–596.
    1. Walker EJ, Su H, Shen F, Choi EJ, Oh SP, Chen G, et al. Arteriovenous malformation in the adult mouse brain resembling the human disease. Ann Neurol. 2011;69(6):954–962. doi: 10.1002/ana.22348.
    1. Han C, Choe SW, Kim YH, Acharya AP, Keselowsky BG, Sorg BS, et al. VEGF neutralization can prevent and normalize arteriovenous malformations in an animal model for hereditary hemorrhagic telangiectasia 2. Angiogenesis. 2014;17(4):823–830. doi: 10.1007/s10456-014-9436-3.
    1. Walker EJ, Su H, Shen F, Degos V, Amend G, Jun K, et al. Bevacizumab attenuates VEGF-induced angiogenesis and vascular malformations in the adult mouse brain. Stroke. 2012;43(7):1925–1930. doi: 10.1161/STROKEAHA.111.647982.
    1. Dupuis-Girod S, Ginon I, Saurin JC, Marion D, Guillot E, Decullier E, et al. Bevacizumab in patients with hereditary hemorrhagic telangiectasia and severe hepatic vascular malformations and high cardiac output. JAMA. 2012;307(9):948–955. doi: 10.1001/jama.2012.250.
    1. Iyer VN, Apala DR, Pannu BS, Kotecha A, Brinjikji W, Leise MD, et al. Intravenous bevacizumab for refractory hereditary hemorrhagic telangiectasia-related epistaxis and gastrointestinal bleeding. Mayo Clin Proc. 2018;93(2):155–166. doi: 10.1016/j.mayocp.2017.11.013.
    1. Dupuis-Girod S, Ambrun A, Decullier E, Fargeton AE, Roux A, Breant V, et al. Effect of bevacizumab nasal spray on epistaxis duration in hereditary hemorrhagic telangectasia: a randomized clinical trial. JAMA. 2016;316(9):934–942. doi: 10.1001/jama.2016.11387.
    1. Riss D, Burian M, Wolf A, Kranebitter V, Kaider A, Arnoldner C. Intranasal submucosal bevacizumab for epistaxis in hereditary hemorrhagic telangiectasia: a double-blind, randomized, placebo-controlled trial. Head Neck. 2015;37(6):783–787. doi: 10.1002/hed.23655.
    1. Azzopardi N, Dupuis-Girod S, Ternant D, Fargeton AE, Ginon I, Faure F, et al. Dose–response relationship of bevacizumab in hereditary hemorrhagic telangiectasia. MAbs. 2015;7(3):630–637. doi: 10.1080/19420862.2015.1022693.
    1. Kim YH, Kim MJ, Choe SW, Sprecher D, Lee YJ. Selective effects of oral antiangiogenic tyrosine kinase inhibitors on an animal model of hereditary hemorrhagic telangiectasia. J Thromb Haemost. 2017;15(6):1095–1102. doi: 10.1111/jth.13683.
    1. Shovlin CL, Guttmacher AE, Buscarini E, Faughnan ME, Hyland RH, Westermann CJ, et al. Diagnostic criteria for hereditary hemorrhagic telangiectasia (Rendu-Osler-Weber syndrome) Am J Med Genet. 2000;91(1):66–67. doi: 10.1002/(SICI)1096-8628(20000306)91:1<66::AID-AJMG12>;2-P.
    1. Hoag JB, Terry P, Mitchell S, Reh D, Merlo CA. An epistaxis severity score for hereditary hemorrhagic telangiectasia. Laryngoscope. 2010;120(4):838–843. doi: 10.1002/lary.20818.
    1. Parambil JG, Woodard TD, Koc ON. Pazopanib effective for bevacizumab-unresponsive epistaxis in hereditary hemorrhagic telangiectasia. Laryngoscope. 2018
    1. Takahashi K, Saishin Y, Saishin Y, King AG, Levin R, Campochiaro PA. Suppression and regression of choroidal neovascularization by the multitargeted kinase inhibitor pazopanib. Arch Ophthalmol. 2009;127(4):494–499. doi: 10.1001/archophthalmol.2009.27.
    1. Kumar R, Knick VB, Rudolph SK, Johnson JH, Crosby RM, Crouthamel MC, et al. Pharmacokinetic-pharmacodynamic correlation from mouse to human with pazopanib, a multikinase angiogenesis inhibitor with potent antitumor and antiangiogenic activity. Mol Cancer Ther. 2007;6(7):2012–2021. doi: 10.1158/1535-7163.MCT-07-0193.
    1. de Jesus-Gonzalez N, Robinson E, Moslehi J, Humphreys BD. Management of antiangiogenic therapy-induced hypertension. Hypertension. 2012;60(3):607–615. doi: 10.1161/HYPERTENSIONAHA.112.196774.
    1. Hurwitz H, Dowlati A, Savage S, Fernando N, Lasalvia S, Whitehead B, et al. Safety, tolerability and pharmacokinetics of oral administration of GW786034 in patients with solid tumors. J Clin Oncol. 2005;23(16 Suppl):3012. doi: 10.1200/jco.2005.23.16_suppl.3012.

Source: PubMed

3
Sottoscrivi