Effect of ATorvastatin On Chronic subdural Hematoma (ATOCH): a study protocol for a randomized controlled trial

Rongcai Jiang, Dong Wang, Wai Sang Poon, Yi Cheng Lu, Xin Gang Li, Shi Guang Zhao, Ren Zhi Wang, Chao You, Xian Rui Yuan, Jian Min Zhang, Hua Feng, Zhou Fei, Xin Guang Yu, Yuan Li Zhao, Jin Hu, De Zhi Kang, Ru Tong Yu, Guo Dong Gao, Xi De Zhu, Tao Sun, Jie He Hao, Xian Zhi Liu, Ning Su, Shu Yuan Yue, Jian Ning Zhang, Rongcai Jiang, Dong Wang, Wai Sang Poon, Yi Cheng Lu, Xin Gang Li, Shi Guang Zhao, Ren Zhi Wang, Chao You, Xian Rui Yuan, Jian Min Zhang, Hua Feng, Zhou Fei, Xin Guang Yu, Yuan Li Zhao, Jin Hu, De Zhi Kang, Ru Tong Yu, Guo Dong Gao, Xi De Zhu, Tao Sun, Jie He Hao, Xian Zhi Liu, Ning Su, Shu Yuan Yue, Jian Ning Zhang

Abstract

Background: Chronic subdural hematoma (CSDH) is a common disease that is more prevalent in older people. Surgical intervention is a safe treatment of choice. However, the recurrence rate is relatively high and the outcome is not always satisfactory among surgically treated patients. It is believed that aberrant angiogenesis and intracapsular inflammation contribute to the development of CSDH. Atorvastatin is reported to promote angiogenesis and suppress inflammation. We have recently shown that atorvastatin is effective to non-surgically reduce and eliminate CSDH with minimal side effects. Here, we report a clinical research trial protocol that is designed to evaluate the therapeutic effects of atorvastatin on CSDH.

Methods/design: We have designed a multi-center, randomized, placebo-controlled, double blind clinical trial for evaluating the efficacy of oral atorvastatin in reducing CSDH. We have so far recruited 96 patients with CT-confirmed or MRI-confirmed CSDHs from 16 medical centers in China. These patients were originally recruited for the Oriental Neurosurgical Evidence-based Study Team (ONET) study. After informed consent is provided, patients are randomized to receive either atorvastatin (oral 20 mg/night for 8 weeks) or placebo (dextrin for 8 weeks); and followed for 16 weeks after the treatment. The primary outcome is the change in hematoma volume at the end of 8-week treatment. Secondary outcomes include: changes in 1) the hematoma volume at the 4(th), 12(th), and 24(th) weeks; 2) Markwalder's Grading Scale and Glasgow Coma Scale (MGS-GCS); 3) Glasgow Outcome Score (GOS) and 4) Activities of Daily Life-the Barthel Index scale (ADL-BI). Safety will be assessed during the study by monitoring adverse events, laboratory tests, electrocardiography (ECG), measurements of vital signs (temperature, pulse, and blood pressure) and body weight.

Discussion: Results of this trial will provide critical information regarding whether atorvastatin is an effective and safe alternative to surgical treatment of CSDH.

Trial registration: ClinicalTrials.gov Identifier--NCT02024373 The date of trial registration: 7 August 2013.

Figures

Fig. 1
Fig. 1
Flow chart to illustrate the design of the trial. ADL-BI, Activities of Daily Life – the Barthel Index scale, AEs adverse events, ECG electrocardiography, GOS Glasgow Outcome Score, IWRS Interactive Web Response System, MGS-GCS Markwalder’s Grading Scale and Glasgow Coma Scale

References

    1. Emich S, et al. The efficacy of dexamethasone on reduction in the reoperation rate of chronic subdural hematoma – the DRESH study: straightforward study protocol for a randomized controlled trial. Trials. 2014;15:6. doi: 10.1186/1745-6215-15-6.
    1. Lee KS, et al. Origin of chronic subdural haematoma and relation to traumatic subdural lesions. Brain Inj. 1998;12(11):901–10. doi: 10.1080/026990598121972.
    1. de Araujo Silva DO, et al. Chronic subdural hematomas and the elderly: surgical results from a series of 125 cases: Old “horses” are not to be shot! Surg Neurol Int. 2012;3:150. doi: 10.4103/2152-7806.104744.
    1. De Bonis P, et al. Antiplatelet/anticoagulant agents and chronic subdural hematoma in the elderly. PLoS One. 2013;8(7):e68732. doi: 10.1371/journal.pone.0068732.
    1. Lee CH, et al. Spontaneous rapid reduction of a large acute subdural hematoma. J Korean Med Sci. 2009;24(6):1224–6. doi: 10.3346/jkms.2009.24.6.1224.
    1. Nakaguchi H, Tanishima T, Yoshimasu N. Factors in the natural history of chronic subdural hematomas that influence their postoperative recurrence. J Neurosurg. 2001;95(2):256–62. doi: 10.3171/jns.2001.95.2.0256.
    1. Santarius T, Hutchinson PJ. Chronic subdural haematoma: time to rationalize treatment? Br J Neurosurg. 2004;18(4):328–32. doi: 10.1080/02688690400004845.
    1. Stippler M, et al. Chronic subdural hematoma patients aged 90 years and older. Neurol Res. 2013;35(3):243–6. doi: 10.1179/1743132813Y.0000000163.
    1. Sun TF, Boet R, Poon WS. Non-surgical primary treatment of chronic subdural haematoma: preliminary results of using dexamethasone. Br J Neurosurg. 2005;19(4):327–33. doi: 10.1080/02688690500305332.
    1. Delgado-Lopez PD, et al. Dexamethasone treatment in chronic subdural haematoma. Neurocirugia (Astur) 2009;20(4):346–59. doi: 10.1016/S1130-1473(09)70154-X.
    1. Dran G, et al. Effectiveness of adjuvant corticosteroid therapy for chronic subdural hematoma: a retrospective study of 198 cases. Neurochirurgie. 2007;53(6):477–82. doi: 10.1016/j.neuchi.2007.09.146.
    1. Berghauser Pont LM, et al. The role of corticosteroids in the management of chronic subdural hematoma: a systematic review. Eur J Neurol. 2012;19(11):1397–403. doi: 10.1111/j.1468-1331.2012.03768.x.
    1. Weigel R, et al. Angiotensin converting enzyme inhibition for arterial hypertension reduces the risk of recurrence in patients with chronic subdural hematoma possibly by an antiangiogenic mechanism. Neurosurgery. 2007;61(4):788–92. doi: 10.1227/01.NEU.0000298907.56012.E8.
    1. Wang D, et al. Effects of atorvastatin on chronic subdural hematoma: a preliminary report from three medical centers. J Neurol Sci. 2014;336(1-2):237–42. doi: 10.1016/j.jns.2013.11.005.
    1. Hohenstein A, et al. Increased mRNA expression of VEGF within the hematoma and imbalance of angiopoietin-1 and -2 mRNA within the neomembranes of chronic subdural hematoma. J Neurotrauma. 2005;22(5):518–28. doi: 10.1089/neu.2005.22.518.
    1. Nagy JA, et al. VEGF-A induces angiogenesis, arteriogenesis, lymphangiogenesis, and vascular malformations. Cold Spring Harb Symp Quant Biol. 2002;67:227–37. doi: 10.1101/sqb.2002.67.227.
    1. Wang D, et al. Membrane neovascularization and drainage of subdural hematoma in a rat model. J Neurotrauma. 2010;27(8):1489–98. doi: 10.1089/neu.2009.1057.
    1. Wada T, et al. Local elevation of the anti-inflammatory interleukin-10 in the pathogenesis of chronic subdural hematoma. Neurosurg Rev. 2006;29(3):242–5. doi: 10.1007/s10143-006-0019-7.
    1. Frati A, et al. Inflammation markers and risk factors for recurrence in 35 patients with a posttraumatic chronic subdural hematoma: a prospective study. J Neurosurg. 2004;100(1):24–32. doi: 10.3171/jns.2004.100.1.0024.
    1. Stanisic M, et al. Chemokines as markers of local inflammation and angiogenesis in patients with chronic subdural hematoma: a prospective study. Acta Neurochir (Wien) 2012;154(1):113–20. doi: 10.1007/s00701-011-1203-2.
    1. Blum A. HMG-CoA reductase inhibitors (statins), inflammation, and endothelial progenitor cells – New mechanistic insights of atherosclerosis. Biofactors. 2014;40(3):295–302. doi: 10.1002/biof.1157.
    1. Sobrino T, et al. Increased levels of circulating endothelial progenitor cells in patients with ischaemic stroke treated with statins during acute phase. Eur J Neurol. 2012;19(12):1539–46. doi: 10.1111/j.1468-1331.2012.03770.x.
    1. Liu Y, et al. Beneficial effects of statins on endothelial progenitor cells. Am J Med Sci. 2012;344(3):220–6. doi: 10.1097/MAJ.0b013e31824998f9.
    1. Zhu JH, et al. Statins contribute to enhancement of the number and the function of endothelial progenitor cells from peripheral blood. Sheng Li Xue Bao. 2004;56(3):357–64.
    1. Dimmeler S, et al. HMG-CoA reductase inhibitors (statins) increase endothelial progenitor cells via the PI 3-kinase/Akt pathway. J Clin Invest. 2001;108(3):391–7. doi: 10.1172/JCI200113152.
    1. Araujo FA, et al. Atorvastatin inhibits inflammatory angiogenesis in mice through down regulation of VEGF, TNF-alpha and TGF-beta1. Biomed Pharmacother. 2010;64(1):29–34. doi: 10.1016/j.biopha.2009.03.003.
    1. Buttmann M, et al. Atorvastatin partially prevents an inflammatory barrier breakdown of cultured human brain endothelial cells at a pharmacologically relevant concentration. J Neurochem. 2007;102(4):1001–8. doi: 10.1111/j.1471-4159.2007.04563.x.
    1. Poulsen FR, et al. Perindopril and residual chronic subdural hematoma volumes six weeks after burr hole surgery: a randomized trial. Clin Neurol Neurosurg. 2014;123:4–8. doi: 10.1016/j.clineuro.2014.05.003.
    1. Lennernas H. Clinical pharmacokinetics of atorvastatin. Clin Pharmacokinet. 2003;42(13):1141–60. doi: 10.2165/00003088-200342130-00005.
    1. Kapsalaki EZ, et al. Spontaneous resolution of acute cranial subdural hematomas. Clin Neurol Neurosurg. 2007;109(3):287–91. doi: 10.1016/j.clineuro.2006.11.005.
    1. Fujimoto K, et al. Predictors of rapid spontaneous resolution of acute subdural hematoma. Clin Neurol Neurosurg. 2014;118:94–7. doi: 10.1016/j.clineuro.2013.11.030.
    1. Marcikic M, et al. Spontaneous resolution of post-traumatic chronic subdural hematoma: case report. Acta Clin Croat. 2010;49(3):331–4.
    1. Miranda LB, et al. Chronic subdural hematoma in the elderly: not a benign disease. J Neurosurg. 2011;114(1):72–6. doi: 10.3171/2010.8.JNS10298.
    1. Labadie EL, Glover D. Physiopathogenesis of subdural hematomas. Part 1: Histological and biochemical comparisons of subcutaneous hematoma in rats with subdural hematoma in man. J Neurosurg. 1976;45(4):382–92. doi: 10.3171/jns.1976.45.4.0382.
    1. Glover D, Labadie EL. Physiopathogenesis of subdural hematomas. Part 2: Inhibition of growth of experimental hematomas with dexamethasone. J Neurosurg. 1976;45(4):393–7. doi: 10.3171/jns.1976.45.4.0393.
    1. Liu JN, et al. Attenuation of airway inflammation by simvastatin and the implications for asthma treatment: is the jury still out? Exp Mol Med. 2014;46:e113. doi: 10.1038/emm.2014.55.
    1. Siddiqui AJ, et al. Rosuvastatin inhibits TIMP-2 and promotes myocardial angiogenesis. Pharmacology. 2014;93(3-4):178–84. doi: 10.1159/000360860.
    1. Criner GJ, et al. Simvastatin for the prevention of exacerbations in moderate-to-severe COPD. N Engl J Med. 2014;370(23):2201–10. doi: 10.1056/NEJMoa1403086.
    1. National Heart, Lung and Blood Institute ARDS Clinical Trials Network et al. Rosuvastatin for sepsis-associated acute respiratory distress syndrome. N Engl J Med. 2014;370(23):2191–200. doi: 10.1056/NEJMoa1401520.
    1. Barnes PJ. Glucocorticoids. Chem Immunol Allergy. 2014;100:311–6. doi: 10.1159/000359984.
    1. Brennan KM, Urschel KL. Recovery of insulin sensitivity in mature horses after a 3 week course of dexamethasone therapy. Equine Vet J. 2014;46(6):718–21. doi: 10.1111/evj.12242.
    1. Soininen K, et al. Muscle symptoms associated with statins: a series of twenty patients. Basic Clin Pharmacol Toxicol. 2006;98(1):51–4. doi: 10.1111/j.1742-7843.2006.pto_193.x.
    1. Tikkanen MJ, et al. Effect of intensive lipid lowering with atorvastatin on cardiovascular outcomes in coronary heart disease patients with mild-to-moderate baseline elevations in alanine aminotransferase levels. Int J Cardiol. 2013;168(4):3846–52. doi: 10.1016/j.ijcard.2013.06.024.
    1. Kalantari S, Naghipour M. Statin therapy and hepatotoxicity: appraisal of the safety profile of atorvastatin in hyperlipidemic patients. Adv Biomed Res. 2014;3:168. doi: 10.4103/2277-9175.139133.

Source: PubMed

3
Abonnere