Aneurysmal Subarachnoid Hemorrhage Trial RandOmizing Heparin

Overview

A Blind-adjudication Multi-center Phase II Randomized Clinical Trial of Continuous Low-dose Intravenous Heparin Therapy in Coiled Low-grade Aneurysmal Subarachnoid Hemorrhage Patients with Significant Hemorrhage Burden. – STUDY IS TEMPORARILY SUSPENDED WITH PLAN TO RESUME SOON. NO SAFETY CONCERNS

Full Title of Study: “A Blind-adjudication Multi-center Phase II Randomized Clinical Trial of Continuous Low-dose Intravenous Heparin Therapy in Coiled Low-grade Aneurysmal Subarachnoid Hemorrhage Patients With Significant Hemorrhage Burden”

Study Type

  • Study Type: Interventional
  • Study Design
    • Allocation: Randomized
    • Intervention Model: Parallel Assignment
    • Primary Purpose: Treatment
    • Masking: Single (Outcomes Assessor)
  • Study Primary Completion Date: January 2023

Detailed Description

The primary objective of this study is to investigate the safety and clinical effect of a continuous low-dose intravenous unfractionated heparin (LDIVH) infusion for the prevention of aneurysmal subarachnoid hemorrhage (aSAH) induced neurocognitive dysfunction and other delayed neurological deficits. Additionally, increased blood and CSF levels of certain inflammatory biomarkers (IL-6, hsCRP, etc) have been correlated to aSAH patients with poor clinical outcomes. Unfractionated heparin (UFH) has known anti-inflammatory actions. As a result, a secondary objective of this study will be to evaluate whether LDIVH can reduce blood and CSF inflammatory biomarkers levels compared to controls and whether there is any association between inflammatory biomarker levels and cognitive outcomes in aSAH.

Interventions

  • Drug: Continuous Low-Dose IV Unfractionated Heparin Infusion
    • Continuous intravenous infusion of a low-dose unfractionated heparin drip

Arms, Groups and Cohorts

  • No Intervention: Control
    • Standard of Care
  • Experimental: LDIVH (Unfractionated Heparin)
    • Continuous Low-Dose IV Unfractionated Heparin Infusion

Clinical Trial Outcome Measures

Primary Measures

  • Montreal Cognitive Assessment (MoCA)
    • Time Frame: 90-day follow-up visit
    • Primary Clinical Outcome Measure- mean score compared between groups
  • Rate of “Major Bleeding” or “Clinically Relevant Non-Major Bleeding”
    • Time Frame: Patients will be followed for the duration of the hospital stay; an expected average of 3 weeks
    • As defined by the International Society of Thrombosis and Heamostasis (ISTH) Primary Safety Outcome Measure-

Secondary Measures

  • Rate of “Major Bleeding”
    • Time Frame: Participants will be followed for the duration of the hospital stay, an expected average of 3 weeks
    • As defined by the International Society of Thrombosis and Haemostasis (ISTH)
  • Rate of Type II Heparin Induced Thrombocytopenia (HIT)
    • Time Frame: Enrollment through 90-day follow-up visit
  • Rate of Deep Venous Thrombosis (DVT) or Pulmonary Embolism (PE)
    • Time Frame: Enrollment through 90-day follow-up visit
  • All Cause – Mortality Rate
    • Time Frame: Enrollment through 90-day follow-up visit
  • Incidence of Any Fever (> 38.3 degrees C; > or = 101.0 degrees F)
    • Time Frame: Participants will be followed for the duration of the hospital stay, an expected average of 3 weeks
  • Incidence of multiple fevers (> 2 episodes)
    • Time Frame: Participants will be followed for the duration of the hospital stay, an expected average of 3 weeks
  • Mean daily fever burden
    • Time Frame: Participants will be followed for the duration of the hospital stay, an expected average of 3 weeks
    • Daily fever burden = Sum of hourly fever burden over 24 hours; Hourly fever burden = Any hourly temperature recording > 37 degrees C – (minus) 37 degrees C; if temperature is less than or equal to 37 degrees C then the hourly fever burden would be zero.
  • Glasgow Coma Score
    • Time Frame: Enrollment, post-enrollment days #6, 10, discharge, and 90-day follow-up visit
  • National Institutes of Health Stroke Scale (NIHSS)
    • Time Frame: Enrollment, post-enrollment days #6, 10, upon discharge from hospital stay an expected average of about 3 weeks after admission, 90-day follow-up visit
  • Montreal Cognitive Assessment (MoCA)
    • Time Frame: Enrollment, post-enrollment days #6, 10, 1 year follow-up
    • Mean between groups and rate of MoCA score of 20 or less between groups
  • Center for Epidemiologic Studies Depression Scale (CES-D)
    • Time Frame: 90-day follow-up visit and 1-year follow-up visit
  • Trail Making Test Parts A&B
    • Time Frame: 90-day follow-up visit
  • Cerebral Vasospasm
    • Time Frame: Participants will be followed for the duration of the hospital stay, an expected average of 3 weeks
    • Incidence of moderate and severe radiographic cerebral vasospasm (catheter angiogram, CTA, MRA) or incidence OR moderate and severe vasospasm by transcranial doppler (TCD) criteria
  • Incidence of clinical cerebral vasospasm requiring rescue therapy
    • Time Frame: Participants will be followed for the duration of the hospital stay, an expected average of 3 weeks
    • Rescue therapy = vasopressors or endovascular therapy for the purposes of reversing clinical vasospasm; it does not include Triple H (Hyperdynamic Therapy)
  • Incidence of CT or MRI imaging demonstrating cerebral vasospasm related cerebral infarction
    • Time Frame: Participants will be followed for the duration of the hospital stay, an expected average of 3 weeks
  • Ordinal Regression Analysis of the modified Rankin Scale score (mRS)
    • Time Frame: 90-day follow-up visit and 1 year follow-up visit
  • Relative frequency of “good outcome” as defined by dichotomized mRS score 0-2
    • Time Frame: 90-day follow-up visit and 1 year follow-up visit
  • Barthel Index
    • Time Frame: 90-day follow-up visit and 1 year follow-up visit
  • Return to work status
    • Time Frame: 90-day follow-up and 1 year follow-up visit
  • Lawton instrumental activities of daily living (IADL)
    • Time Frame: 90-day follow-up visit and 1 year follow-up visit
  • Quality of Life in Brain Injury – Overall Scale (QOLIBRI-OS)
    • Time Frame: 90-day follow-up visit and 1 year follow-up visit
  • Checklist Individual Strength- Subscale Fatigue (CIS-F)
    • Time Frame: 90-day follow-up visit and 1 year follow-up visit
  • Plasma biomarker level (hsCRP)
    • Time Frame: Enrollment, post-enrollment days #2,4,6,10
  • Cerebrospinal Fluid (CSF) biomarker level (hsCRP)
    • Time Frame: Enrollment, post-enrollment days #2,4,6,10
  • Rate of Serious Adverse Events (SAEs)
    • Time Frame: From enrollment through 90-Day follow-up visit
    • Secondary Safety Outcome Measure

Participating in This Clinical Trial

Inclusion Criteria

1. Age ≥ 18 and ≤ 70 years 2. Historical modified Rankin Scale Score 0-1 3. Aneurysmal subarachnoid hemorrhage caused by a ruptured saccular aneurysm confirmed by catheter angiography that is repaired by endovascular coil embolization. Initiation of the coil embolization procedure should occur within 48 hours from the time of the aneurysm rupture (ictus). In patients where the exact time of the ictus is uncertain, a reasonable estimate of the time of ictus may be assigned. This reasonable time estimate should be considered likely accurate to within hours of the true unknown time. 4. Quality of aneurysm embolization is interpreted to be Raymond-Roy Score of 1 (Complete) or 2 (Residual Neck) indicating that the aneurysm is adequately secured. A tiny amount of contrast in the body of the aneurysm is acceptable as long as the physician considers the aneurysm secured and to NOT represent a Raymond-Roy Score of 3 (Residual Aneurysm). 5. WFNS grade 1 or 2 as assessed after repair of the aneurysm during screening but prior to randomization. A patient who presents with a WFNS greater than 2 who then improves with resuscitation, ventriculostomy, or time is acceptable. 6. The pre-repair, admission head CT demonstrates an aSAH bleed pattern of "thick and diffuse" or "thick and focal" hemorrhage within the subarachnoid basal cisterns measuring ≥ 4 mm in the short axis and ≥ 20 mm in the long axis which is consistent with a modified Fisher grade 3 or 4. Intraventricular hemorrhage is acceptable. Enrollable patients must NOT have a parenchymal hemorrhage greater than 10 cc. Please refer to diagram below for examples. The hemorrhage location should be substantially within the supratentorial space and not isolated to the infratentorial space. 7. The location of the aneurysm should be the anterior circulation, posterior communicating, OR a basilar terminus (apex). Angiographic location of the aneurysm should be confirmed by catheter digital subtraction angiography (DSA) usually obtained during the coil embolization procedure. Patients with PICA or other posterior circulation aneurysms as the cause of the SAH should not be included because they typically cause primarily infratentorial bleed patterns. 8. Ability to screen the patient and obtain a head CT 2-12 hours after the completion of the coiling procedure and the ability to initiate the study drug 12 ± 8 hours after the completion of aneurysm coiling procedure. 9. After recovering from anesthesia following the aneurysm coiling procedure, the patient must remain a WFNS SAH grade ≤ 2 without evidence of a significant new focal neurological deficit including monoparesis / monoplegia, hemiparesis / hemiplegia, or receptive, expressive or global aphasia. New minor cranial nerve defect without any other new findings is permissible. If an NIHSS score was obtained prior to the aneurysm coiling procedure, a post-coiling (pre-enrollment) NIHSS score must not have increased by ≥ 4 points and GCS score must not be decreased by ≥ 2 points. The clinician at the local site should use their best clinical judgment as to whether a significant neurological decline has occurred due to the procedure. 10. Patient is willing and able to return for study follow-up visits. 11. Patient or their Legally Authorized Representative (LAR) has provided written informed consent. Exclusion Criteria:

1. Angio-negative SAH. 2. History or imaging suggesting that the current hemorrhage presentation is a recent re-rupture of the aneurysm. Prior sentinel headache with negative CT or prior sentinel headache where the patient did not seek medical attention does not exclude the patient. 3. Surgical Clipping (or plan for clipping) of the ruptured aneurysm or any non- ruptured aneurysm on the same admission. 4. Aneurysm is identified to be traumatic, mycotic, blister or fusiform type by catheter DSA. 5. Any intracranial stent placement or non-coil intra-aneurysmal device where dual- antiplatelet therapy is needed during admission. 6. Patient has additional aneurysm(s) that are untreated and could reasonably be considered a possible alternate cause of the aSAH based on the observed bleeding pattern. Adequate treatment of these aneurysms by coiling embolization would result in the aneurysms no longer causing an exclusion. MRI may be used in some situations to determine that the associated aneurysms did not rupture based on lack of blood seen adjacent to the additional aneurysms. 7. Patient received heparin in any form within the last 100 days prior to current presentation / admission. 8. Thrombocytopenia (platelet count less than 100,000 – assuming clumping has been ruled out as a cause). 9. New intraparenchymal hemorrhage or new infarction larger the 15cc in volume, or significant increased mass effect as seen on the post-coiling, pre-enrollment head CT when compared to baseline admission head CT. New hyperdensity on CT scan related to contrast staining is not an exclusion. 10. Patient has a documented history of heparin induced thrombocytopenia (HIT). 11. Patient developed SAH-induced cardiac stunning prior to enrollment, with an ejection fraction <30%, or requiring IV medications for blood pressure maintenance. 12. Concurrent significant intracranial pathology identified prior to enrollment, including but not limited to, Moyamoya disease, high suspicion or documented CNS vasculitis, severe fibromuscular dysplasia, arteriovenous malformation, arteriovenous fistula, or malignant brain tumor. 13. Thrombolytic therapy within 24 hours prior to enrollment (rtPA, urokinase, etc.) 14. Plan for antiplatelet or oral anticoagulation therapy from the time of the coil embolization procedure until 14 full days after enrollment. Antiplatelet therapy may be resumed after the 14-day window. A single 325 mg Aspirin (or lower dose) given during the coil embolization peri-procedural period is acceptable if this is the local standard of care but should be documented. 15. Concomitant serious or uncontrolled disease such as severe infection, active (non- remission) cancer, severe organ dysfunction (severe heart failure, severe chronic kidney impairment requiring dialysis or severe chronic liver disease) or any coagulopathy (including DIC or bleeding diathesis). 16. Uncontrollable hypertension (>180 systolic and/or >110 diastolic) that is not correctable prior to enrollment. 17. Prior neurological disease/deficit or psychiatric disease that may continue to alter the results of neuropsychological evaluation, such as dementia, Multiple sclerosis, seizure disorder, severe traumatic brain injury, previous ruptured cerebral aneurysm or active major depression. Childhood seizures that have resolved and no longer require treatment are not part of this exclusion criteria. 18. Active Immunosuppression therapy including chronic corticosteroid usage. 19. History of gastrointestinal hemorrhage or major systemic hemorrhage within 30 days (including large flank or large retroperitoneal hematoma due to current admission coiling procedure requiring treatment), hemoglobin less than 6 g/dL, INR ≥1.5 after reversal of anticoagulants. 20. Major surgery (including open femoral, aortic, or carotid surgery) within previous 30 days. 21. Currently pregnant. 22. Enrollment in another research study that prescribes a therapeutic treatment that differs from the local standard of care, or that would conflict with this study in some other significant fashion. Registries or coil comparison studies are appropriate.

Gender Eligibility: All

Minimum Age: 18 Years

Maximum Age: 70 Years

Are Healthy Volunteers Accepted: No

Investigator Details

  • Lead Sponsor
    • Robert F. James
  • Collaborator
    • Indiana University School of Medicine
  • Provider of Information About this Clinical Study
    • Sponsor-Investigator: Robert F. James, Professor of Neurosurgery – Indiana University
  • Overall Official(s)
    • Robert F James, MD, Principal Investigator, Indiana University
    • J Marc Simard, MD, PhD, Principal Investigator, University of Maryland
    • J Mocco, MD, MSc, Principal Investigator, Icahn School of Medicine at Mount Sinai
    • Kevin N Sheth, MD, Principal Investigator, Yale University

References

Simard JM, Schreibman D, Aldrich EF, Stallmeyer B, Le B, James RF, Beaty N. Unfractionated heparin: multitargeted therapy for delayed neurological deficits induced by subarachnoid hemorrhage. Neurocrit Care. 2010 Dec;13(3):439-49. doi: 10.1007/s12028-010-9435-1.

Al-Khindi T, Macdonald RL, Schweizer TA. Cognitive and functional outcome after aneurysmal subarachnoid hemorrhage. Stroke. 2010 Aug;41(8):e519-36. doi: 10.1161/STROKEAHA.110.581975. Epub 2010 Jul 1.

Schweizer TA, Al-Khindi T, Macdonald RL. Mini-Mental State Examination versus Montreal Cognitive Assessment: rapid assessment tools for cognitive and functional outcome after aneurysmal subarachnoid hemorrhage. J Neurol Sci. 2012 May 15;316(1-2):137-40. doi: 10.1016/j.jns.2012.01.003. Epub 2012 Jan 26.

Wong GK, Lam SW, Ngai K, Wong A, Mok V, Poon WS. Quality of Life after Brain Injury (QOLIBRI) Overall Scale for patients after aneurysmal subarachnoid hemorrhage. J Clin Neurosci. 2014 Jun;21(6):954-6. doi: 10.1016/j.jocn.2013.09.010. Epub 2013 Nov 9.

Hong CM, Tosun C, Kurland DB, Gerzanich V, Schreibman D, Simard JM. Biomarkers as outcome predictors in subarachnoid hemorrhage–a systematic review. Biomarkers. 2014 Mar;19(2):95-108. doi: 10.3109/1354750X.2014.881418. Epub 2014 Feb 5.

Romero FR, Bertolini Ede F, Figueiredo EG, Teixeira MJ. Serum C-reactive protein levels predict neurological outcome after aneurysmal subarachnoid hemorrhage. Arq Neuropsiquiatr. 2012 Mar;70(3):202-5. doi: 10.1590/s0004-282×2012000300009.

Fountas KN, Tasiou A, Kapsalaki EZ, Paterakis KN, Grigorian AA, Lee GP, Robinson JS Jr. Serum and cerebrospinal fluid C-reactive protein levels as predictors of vasospasm in aneurysmal subarachnoid hemorrhage. Clinical article. Neurosurg Focus. 2009 May;26(5):E22. doi: 10.3171/2009.2.FOCUS08311.

Tosun C, Kurland DB, Mehta R, Castellani RJ, deJong JL, Kwon MS, Woo SK, Gerzanich V, Simard JM. Inhibition of the Sur1-Trpm4 channel reduces neuroinflammation and cognitive impairment in subarachnoid hemorrhage. Stroke. 2013 Dec;44(12):3522-8. doi: 10.1161/STROKEAHA.113.002904. Epub 2013 Oct 10.

Simard JM, Aldrich EF, Schreibman D, James RF, Polifka A, Beaty N. Low-dose intravenous heparin infusion in patients with aneurysmal subarachnoid hemorrhage: a preliminary assessment. J Neurosurg. 2013 Dec;119(6):1611-9. doi: 10.3171/2013.8.JNS1337. Epub 2013 Sep 13.

James RF, Shao EY, Page PS, Nazar RG, Martin LB, Dvorak J, Kanaan HK, Daniels MJ, Craycroft J, Rai SN, Everhart DE, Simard JM. Low-dose IV heparin preserves cognitive function in aneurysmal subarachnoid hemorrhage patients. [Unpublished Data]. Presented at AANS 82nd Annual Scientific Meeting. April 5-9, 2014. San Francisco, CA; Abstract #16572.

Clinical trials entries are delivered from the US National Institutes of Health and are not reviewed separately by this site. Please see the identifier information above for retrieving further details from the government database.

At TrialBulletin.com, we keep tabs on over 200,000 clinical trials in the US and abroad, using medical data supplied directly by the US National Institutes of Health. Please see the About and Contact page for details.