search
Back to results

Effect of Xenon on Brain Injury After Aneurysmal Subarachnoid Hemorrhage (Xe-SAH)

Primary Purpose

Subarachnoid Hemorrhage, Aneurysmal, Cerebral Injury, Cerebral Ischemia

Status
Not yet recruiting
Phase
Phase 2
Locations
International
Study Type
Interventional
Intervention
Xenon
air/oxygen
Sponsored by
Turku University Hospital
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Subarachnoid Hemorrhage, Aneurysmal focused on measuring xenon, neuroprotection, aneurysmal subarachnoid hemorrhage

Eligibility Criteria

18 Years - undefined (Adult, Older Adult)All SexesDoes not accept healthy volunteers

Inclusion Criteria:

To be considered eligible to participate in this study, a SAH subject must meet the inclusion criteria listed below:

  1. Informed consent obtained from the next of kin or legal representative
  2. Aneurysmal subarachnoid hemorrhage visible on CTA or DSA.
  3. Deterioration of consciousness to Hunt-Hess 3-5
  4. Age of ≥ 18 years
  5. Intubated.
  6. GCS 3-12 obtained off neuromuscular blocking agents
  7. Xenon treatment can be started within 6 hours after onset of SAH symptoms

Exclusion Criteria:

An aSAH subject may not be enrolled in the trial if he/she meets any one of the exclusion criteria below:

  1. Acute or chronic traumatic brain injury
  2. Maximum diameter of intracerebral hemorrhage > 2.5 cm
  3. Pneumothorax or pneumomediastinum,
  4. Acute lung injury requiring ≥ 60% FIO2 (fraction of inspired oxygen).
  5. Systolic arterial pressure < 80 mmHg or mean arterial pressure < 60 mmHg for over 30 min period
  6. Bilaterally fixed and dilated pupils
  7. Positive pregnancy test, known pregnancy, or current breast-feeding
  8. Neurological deficiency due to traumatic brain injury or other neurological illness
  9. Imminent death or current life-threatening disease
  10. Current enrollment in another interventional study
  11. The subject is known to have clinically significant laboratory abnormality, medical condition (such as decompensated liver disease or severe chronic obstructive pulmonary disease), or social circumstance that, in the investigator's opinion, makes it inappropriate for the subject to participate in this clinical trial.
  12. Presence of implants or foreign bodies which are not known to be MRI safe

Sites / Locations

  • Aalto University School of Science
  • Kuopio University Hospital
  • Tampere University Hospital
  • Turku University Hospital
  • Elomatic
  • University of Turku, Turku Bioscience, Analysis of the metabolomics
  • Örebro University

Arms of the Study

Arm 1

Arm 2

Arm Type

Active Comparator

Experimental

Arm Label

Air/Oxygen

xenon

Arm Description

Control arm: air/oxygen with standard of care

Xenon arm: xenon inhalation in air/oxygen with standard of care

Outcomes

Primary Outcome Measures

Fractional anisotropy of the white matter
Global fractional anisotropy of white matter of diffusion tensor imaging (DTI). Hypothesis: White matter damage is less severe in xenon treated patients, i.e. global fractional anisotropy is significantly higher in the xenon group than in the control group as assessed with the 1st MRI.

Secondary Outcome Measures

Fractional anisotropy of white matter at cerebellum and/or at corpus callosum as assessed with the 1st MRI.
Fractional anisotropy of white matter at cerebellum and/or at corpus callosum as assessed with the 1st MRI.
Safety and tolerability of xenon
Safety and tolerability of xenon as assessed with a ratio of adverse events, serious adverse events and suspected unexpected serious adverse reactions (SUSARs) during the follow-up of one year between the xenon group and the control group.
Composite of radiological early brain injury (EBI) and delayed cerebral ischemia (DCI)
Composite of radiological EBI (within 72 hours after start of SAH symptoms) and DCI (Criterion of DCI: 1. a new focal neurological deficit (such as hemiparesis, aphasia, apraxia, hemianopia, or neglect) /decrease in level of consciousness (i.e. decrease of at least 2 points on the Glasgow Coma Scale; either on the total score or on one of its individual components, such as eye, motor on either side, or verbal). This should last for at least 1 hour and not is due to other causes (e.g. hydrocephalus, seizures, metabolic derangement, infection, sedation) and is not apparent immediately after aneurysm occlusion, and cannot be attributed to other causes by means of clinical assessment, CT or MRI scanning of the brain, and appropriate laboratory studies, 2. a new infarct on follow-up imaging (i.e. in any of the following: 2nd MRI, CT, CTA, DSA and perfusion CT) after 4 days post-SAH, or 3. both 1 and 2), and poor outcome at 3-months (good: mRS 0-2; poor: mRS 3-6) at 3-months and at 1 year
Neurogenic Stress Cardiomyopathy and Stunned Myocardium
Neurogenic Stress Cardiomyopathy and Stunned Myocardium (i.e. myocardial injury caused by sympathetic storm and autonomic dysregulation with hs-troponin elevation, left ventricular dysfunction or ECG changes)
Intracerebral pressure (ICP)
ICP level Duration of therapy for ICP control/monitoring
Intracerebral pressure (ICP)
Need for ICP therapies (hypothermia, decompressive craniotomy)
Intracerebral pressure (ICP)
Duration of therapy for ICP control/monitoring
Plasma catecholamine level
Plasma level of noradrenaline , adrenaline, and dopamine
Selected biomarkers
Selected biomarkers of brain injury: neurofilament light (NF-L), glial fibrillary acidic protein (GFAP), calcium binding protein S100B (S100B), ubiquitin carboxyterminal hydrolase L1 (UCH-L1), total tau, cytokines (tumour necrosis factor alpha, interleukins 6 and 10)
Development of prognostication models
Development of prognostication models with a selected combination of brain imaging, clinical data, biomarkers and metabolomics by applying artificial intelligence and machine learning for long-term outcome after aSAH
Development of prognostication models
Development of prognostication models with a selected combination of brain imaging, clinical data, biomarkers and metabolomics by applying artificial intelligence and machine learning for DCI after aSAH
Development of prognostication models
Development of prognostication models with a selected combination of brain imaging, clinical data, biomarkers and metabolomics by applying artificial intelligence and machine learning for vasospasm after aSAH
Development of prognostication models
Development of prognostication models with a selected combination of brain imaging, clinical data, biomarkers and metabolomics by applying artificial intelligence and machine learning for EBI after aSAH
Difference of MRI parameters between xenon and control group
Difference of MRI parameters (fractional anisotropy, axial diffucivity, radial diffucivity of diffusion tensor imaging, DTI) between xenon and control group and in predicting risk for EBI
Difference of MRI parameters between xenon and control group
Difference of MRI parameters (fractional anisotropy, axial diffucivity, radial diffucivity of DTI) between xenon and control group and in predicting risk for vasospasm
Difference of MRI parameters between xenon and control group
Difference of MRI parameters (fractional anisotropy, axial diffucivity, radial diffucivity of DTI) between xenon and control group and in predicting risk for DCI
Difference of MRI parameters between xenon and control group
Difference of MRI parameters (fractional anisotropy, axial diffucivity, radial diffucivity of DTI) between xenon and control group and in predicting risk for good/poor neurological outcome at 3 moths, at 1 year and at 2 years after onset of aSAH symptoms (mRS 0-2/mRS 3-6).
Difference of CTA findings
Difference of CTA ischemic findings between xenon and control group and in predicting risk for EBI
Difference of CTA findings
Difference of ischemic findings in CTA between xenon and control group and in predicting risk for vasospasm
Difference of CTA findings
Difference of ischemic findings in CTA between xenon and control group and in predicting risk for DCI
Difference of CTA findings between xenon and control group
Difference of ischemic findings in CTA between xenon and control group and in predicting risk for good/poor neurological outcome at 3 moths, at 1 year and at 2 years after onset of aSAH symptoms (mRS 0-2/mRS 3-6).
Difference of DSA findings between xenon and control group
Difference of DSA findings indicating ischemic pattern of perfusion between xenon and control group and in predicting risk for EBI
Difference of DSA findings between xenon and control group
Difference of DSA findings indicating ischemic pattern of perfusion between xenon and control group and in predicting risk for vasospasm
Difference of DSA findings between xenon and control group
Difference of DSA findings indicating ischemic pattern of perfusion between xenon and control group and in predicting risk for DCI
Difference of DSA findings between xenon and control group
Difference of DSA findings indicating ischemic pattern of perfusion between xenon and control group and in predicting risk for good/poor neurological outcome at 3 moths, at 1 year and at 2 years after onset of aSAH symptoms (mRS 0-2/mRS 3-6).
Activity of microglia cells assessed with PET
It will be explored whether [11C](R)-PK11195 can be used to test the hypothesis of neuroprotective effect of xenon and to explore the role of inflammatory process for DCI after SAH. This could be demonstrated by showing less microglial activation in xenon group than in the reference therapy group and in the patients with good outcome, i.e. no DCI; Difference of activity of microglia cells between xenon and control group and in predicting risk for DCI
Activity of microglia cells assessed with PET
It will be explored whether [11C](R)-PK11195 can be used to test the hypothesis of neuroprotective effect of xenon and to explore the role of inflammatory process for neurological outcome after SAH. This could be demonstrated by showing less microglial activation in xenon group than in the reference therapy group and in the patients with good outcome, i.e. mRS 0-2;
Cerebral fluid dynamics
Predictive value of CFD simulations assessed with 3 dimensional DSA within 4 days of ICU arrival in predicting risk for EBI within 72 hours after onset of aSAH symptoms
Cerebral fluid dynamics
Predictive value of CFD simulations assessed with 3 dimensional DSA within 21 days of ICU arrival in predicting risk for neurological outcome at 3 months, at 1 year and at 2 years after SAH (mRS 0-2)
Cerebral fluid dynamics
Predictive value of CFD simulations assessed with 3 dimensional DSA within 21 days of ICU arrival in predicting risk for DCI within 6 weeks after onset of aSAH symptoms

Full Information

First Posted
January 2, 2021
Last Updated
April 18, 2023
Sponsor
Turku University Hospital
Collaborators
Academy of Finland
search

1. Study Identification

Unique Protocol Identification Number
NCT04696523
Brief Title
Effect of Xenon on Brain Injury After Aneurysmal Subarachnoid Hemorrhage
Acronym
Xe-SAH
Official Title
Effect of Xenon on Brain Injury, Neurological Outcome and Survival in Patients After Aneurysmal Subarachnoid Hemorrhage
Study Type
Interventional

2. Study Status

Record Verification Date
April 2023
Overall Recruitment Status
Not yet recruiting
Study Start Date
October 1, 2023 (Anticipated)
Primary Completion Date
December 31, 2026 (Anticipated)
Study Completion Date
December 31, 2027 (Anticipated)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Principal Investigator
Name of the Sponsor
Turku University Hospital
Collaborators
Academy of Finland

4. Oversight

Studies a U.S. FDA-regulated Drug Product
No
Studies a U.S. FDA-regulated Device Product
No
Data Monitoring Committee
Yes

5. Study Description

Brief Summary
An investigator-initiated clinical drug study Main Objective: To explore neuroprotective properties of xenon in patients after aneurysmal subarachnoid hemorrhage (SAH). Primary endpoint: Global fractional anisotropy of white matter of diffusion tensor imaging (DTI). Hypothesis: White matter damage is less severe in xenon treated patients, i.e. global fractional anisotropy is significantly higher in the xenon group than in the control group as assessed with the 1st magnetic resonance imaging (MRI). After confirmation of aSAH and obtaining a signed assent subjects will be randomized to the following groups: Control group: Standard of Care (SOC) group: Air/oxygen and Normothermia 36.5-37.5°C; Xenon group: Normothermia 36.5-37.5°C +Xenon inhalation in air/oxygen for 24 hours. Brain magnetic resonance imaging techniques will be undertaken to evaluate the effects of the intervention on white and grey matter damage and neuronal loss. Neurological outcome will be evaluated at 3, 12 and 24 months after onset of aSAH symptoms Investigational drug/treatment, dose and mode of administration: 50±2 % end tidal concentration of inhaled xenon in oxygen/air. Comparative drug(s)/placebo/treatment, dose and mode of administration: Standard of care treatment according to local and international consensus reports. Duration of treatment: 24 hours Assessments: Baseline data Information that characterizes the participant's condition prior to initiation of experimental treatment is obtained as soon as is clinically reasonable. These include participant demographics, medical history, vital signs, oxygen saturation, and concentration of oxygen administered. Acute data The collected information will contain quantitative and qualitative data of aSAH patients, as recommended by recent recommendations of the working group on subject characteristics, and including all relevant Common Data Elements (CDE) can be applied. Specific definitions, measurements tools, and references regarding each SAH CDE can be found on the weblink here: https://www.commondataelements.ninds.nih.gov/SAH.aspx#tab=Data_Standards.
Detailed Description
Assessments of efficacy: A brain Computer tomography angiography (CTA) and / or 3 D Digital subtraction angiography (DSA) (whenever possible instead of 2D DSA) will be performed at hospital arrival and whenever clinically indicated. 1st 3 Tesla MRI 72 ± 24 hours after onset of aSAH symptoms; 2nd 3 Tesla MRI 42 ± 4 days after onset of aSAH symptoms. 3D DSA: Computational fluid dynamic simulations (CFD), artificial intelligence and machine learning. Brain Positron emission tomography (PET): The 1st 4 ± 1 weeks and the 2nd at 3 months after onset of aSAH symptoms. Biochemical assessment: A blood samples of 20 ml for determination of plasma catecholamines, plasma metabolomics (see details of metabolomics in section 18.4.7), cardiac enzyme release (P-hs-troponin-T and heart fatty-acid binding protein), selected biomarkers will be analysed at intensive crae unit (ICU) arrival and at 24h, at 48h and at 72h after onset of SAH symptoms. In addition, a sample of spinal fluid will be collected through external ventricular drainage (EVD) at ICU arrival or as soon as it is in place and at 24h, at 48h and at 72h after onset of SAH symptoms for assessment of metabolomics Electrocardiograph (ECG) at ICU arrival and at 24h, at 48h and at 72h after onset of aSAH symptoms. Neurological evaluation: at 3, 12 and at 24 months after aSAH with GOSe, Modified ranking score (mRS). Statistical methods: 1) Basic statistical tests (t-tests, Mann-Whitney, Chi square, etc); 2) Survival analysis methods; 3) An analysis of variance for repeated measurements; 4) A sample size of 100 is estimated on the basis of a recent studies in SAH patients to provide 80% power with a 2-sided α level of 0.05 to detect a mean difference of 0.02 (SD 0.035) in the global fractional anisotropy of white matter between the xenon group and the control group (98). Accordingly, this mean difference is estimated to have a predictive value for DCI and poor neurological outcome (i.e. mRS 3-6).Significance level of 0.05 and an estimation of 95 % confidence intervals will be used in the statistical analyses.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Subarachnoid Hemorrhage, Aneurysmal, Cerebral Injury, Cerebral Ischemia, Cerebral Infarction, Cardiac Event, Cardiac Failure
Keywords
xenon, neuroprotection, aneurysmal subarachnoid hemorrhage

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 2
Interventional Study Model
Parallel Assignment
Model Description
Study design is a single blind randomized two-armed parallel follow-up study.
Masking
ParticipantOutcomes Assessor
Masking Description
single blind; participants, outcomes assessors are blinded
Allocation
Randomized
Enrollment
160 (Anticipated)

8. Arms, Groups, and Interventions

Arm Title
Air/Oxygen
Arm Type
Active Comparator
Arm Description
Control arm: air/oxygen with standard of care
Arm Title
xenon
Arm Type
Experimental
Arm Description
Xenon arm: xenon inhalation in air/oxygen with standard of care
Intervention Type
Drug
Intervention Name(s)
Xenon
Intervention Description
Xenon arm will be treated with xenon inhalation with endtidal concentration of 50 % in air/oxygen and with standard of care
Intervention Type
Drug
Intervention Name(s)
air/oxygen
Intervention Description
Control group will be treated with air/oxygen
Primary Outcome Measure Information:
Title
Fractional anisotropy of the white matter
Description
Global fractional anisotropy of white matter of diffusion tensor imaging (DTI). Hypothesis: White matter damage is less severe in xenon treated patients, i.e. global fractional anisotropy is significantly higher in the xenon group than in the control group as assessed with the 1st MRI.
Time Frame
48-96 hours after start of aSAH symptoms
Secondary Outcome Measure Information:
Title
Fractional anisotropy of white matter at cerebellum and/or at corpus callosum as assessed with the 1st MRI.
Description
Fractional anisotropy of white matter at cerebellum and/or at corpus callosum as assessed with the 1st MRI.
Time Frame
48-96 hours after start of aSAH symptoms
Title
Safety and tolerability of xenon
Description
Safety and tolerability of xenon as assessed with a ratio of adverse events, serious adverse events and suspected unexpected serious adverse reactions (SUSARs) during the follow-up of one year between the xenon group and the control group.
Time Frame
during the follow-up of one year
Title
Composite of radiological early brain injury (EBI) and delayed cerebral ischemia (DCI)
Description
Composite of radiological EBI (within 72 hours after start of SAH symptoms) and DCI (Criterion of DCI: 1. a new focal neurological deficit (such as hemiparesis, aphasia, apraxia, hemianopia, or neglect) /decrease in level of consciousness (i.e. decrease of at least 2 points on the Glasgow Coma Scale; either on the total score or on one of its individual components, such as eye, motor on either side, or verbal). This should last for at least 1 hour and not is due to other causes (e.g. hydrocephalus, seizures, metabolic derangement, infection, sedation) and is not apparent immediately after aneurysm occlusion, and cannot be attributed to other causes by means of clinical assessment, CT or MRI scanning of the brain, and appropriate laboratory studies, 2. a new infarct on follow-up imaging (i.e. in any of the following: 2nd MRI, CT, CTA, DSA and perfusion CT) after 4 days post-SAH, or 3. both 1 and 2), and poor outcome at 3-months (good: mRS 0-2; poor: mRS 3-6) at 3-months and at 1 year
Time Frame
EBI: within first 72 hours after start of aSAH symptoms; mRS at 3 months and at 1 year and at 2 years after onset of aSAH symptoms
Title
Neurogenic Stress Cardiomyopathy and Stunned Myocardium
Description
Neurogenic Stress Cardiomyopathy and Stunned Myocardium (i.e. myocardial injury caused by sympathetic storm and autonomic dysregulation with hs-troponin elevation, left ventricular dysfunction or ECG changes)
Time Frame
follow-up of 1 year
Title
Intracerebral pressure (ICP)
Description
ICP level Duration of therapy for ICP control/monitoring
Time Frame
during ICU stay up to 14 days after onset of aSAH symptoms
Title
Intracerebral pressure (ICP)
Description
Need for ICP therapies (hypothermia, decompressive craniotomy)
Time Frame
during ICU stay up to 14 days after onset of aSAH symptoms
Title
Intracerebral pressure (ICP)
Description
Duration of therapy for ICP control/monitoring
Time Frame
during ICU stay up to 14 days after onset of aSAH symptoms
Title
Plasma catecholamine level
Description
Plasma level of noradrenaline , adrenaline, and dopamine
Time Frame
within 3 hours of ICU arrival, at 24h, 48h and 72 h after onset of aSAH symptoms
Title
Selected biomarkers
Description
Selected biomarkers of brain injury: neurofilament light (NF-L), glial fibrillary acidic protein (GFAP), calcium binding protein S100B (S100B), ubiquitin carboxyterminal hydrolase L1 (UCH-L1), total tau, cytokines (tumour necrosis factor alpha, interleukins 6 and 10)
Time Frame
within 3 hours of ICU arrival and at 24h, at 48h and at 72h after onset of aSAH symptoms
Title
Development of prognostication models
Description
Development of prognostication models with a selected combination of brain imaging, clinical data, biomarkers and metabolomics by applying artificial intelligence and machine learning for long-term outcome after aSAH
Time Frame
long-term outcome at 3 months, at 1 and at 2 years after onset of aSAH symptoms
Title
Development of prognostication models
Description
Development of prognostication models with a selected combination of brain imaging, clinical data, biomarkers and metabolomics by applying artificial intelligence and machine learning for DCI after aSAH
Time Frame
between day 4 and 6 weeks after onset of aSAH symtoms
Title
Development of prognostication models
Description
Development of prognostication models with a selected combination of brain imaging, clinical data, biomarkers and metabolomics by applying artificial intelligence and machine learning for vasospasm after aSAH
Time Frame
within 21 days after onset of aSAH symptoms
Title
Development of prognostication models
Description
Development of prognostication models with a selected combination of brain imaging, clinical data, biomarkers and metabolomics by applying artificial intelligence and machine learning for EBI after aSAH
Time Frame
within 72 hours after onset of aSAH symtoms
Title
Difference of MRI parameters between xenon and control group
Description
Difference of MRI parameters (fractional anisotropy, axial diffucivity, radial diffucivity of diffusion tensor imaging, DTI) between xenon and control group and in predicting risk for EBI
Time Frame
within 72 hours after onset of aSAH symptoms
Title
Difference of MRI parameters between xenon and control group
Description
Difference of MRI parameters (fractional anisotropy, axial diffucivity, radial diffucivity of DTI) between xenon and control group and in predicting risk for vasospasm
Time Frame
within 21 days after onset of aSAH symptoms
Title
Difference of MRI parameters between xenon and control group
Description
Difference of MRI parameters (fractional anisotropy, axial diffucivity, radial diffucivity of DTI) between xenon and control group and in predicting risk for DCI
Time Frame
between day 4 and 6 weeks after onset of aSAH symptoms
Title
Difference of MRI parameters between xenon and control group
Description
Difference of MRI parameters (fractional anisotropy, axial diffucivity, radial diffucivity of DTI) between xenon and control group and in predicting risk for good/poor neurological outcome at 3 moths, at 1 year and at 2 years after onset of aSAH symptoms (mRS 0-2/mRS 3-6).
Time Frame
at 3 months, at 1 year and at 2 years after onset of aSAH symptoms
Title
Difference of CTA findings
Description
Difference of CTA ischemic findings between xenon and control group and in predicting risk for EBI
Time Frame
within 72 hours after onset of aSAH symptoms
Title
Difference of CTA findings
Description
Difference of ischemic findings in CTA between xenon and control group and in predicting risk for vasospasm
Time Frame
within 21 days after onset of aSAH symptoms
Title
Difference of CTA findings
Description
Difference of ischemic findings in CTA between xenon and control group and in predicting risk for DCI
Time Frame
between day 4 and 6 weeks after onset of aSAH symptoms
Title
Difference of CTA findings between xenon and control group
Description
Difference of ischemic findings in CTA between xenon and control group and in predicting risk for good/poor neurological outcome at 3 moths, at 1 year and at 2 years after onset of aSAH symptoms (mRS 0-2/mRS 3-6).
Time Frame
at 3 months, at 1 year and at 2 years after onset of aSAH symptoms
Title
Difference of DSA findings between xenon and control group
Description
Difference of DSA findings indicating ischemic pattern of perfusion between xenon and control group and in predicting risk for EBI
Time Frame
within 72 hours after onset of aSAH symptoms
Title
Difference of DSA findings between xenon and control group
Description
Difference of DSA findings indicating ischemic pattern of perfusion between xenon and control group and in predicting risk for vasospasm
Time Frame
within 21 days after onset of aSAH symptoms
Title
Difference of DSA findings between xenon and control group
Description
Difference of DSA findings indicating ischemic pattern of perfusion between xenon and control group and in predicting risk for DCI
Time Frame
between day 4 and 6 weeks after onset of aSAH symptoms
Title
Difference of DSA findings between xenon and control group
Description
Difference of DSA findings indicating ischemic pattern of perfusion between xenon and control group and in predicting risk for good/poor neurological outcome at 3 moths, at 1 year and at 2 years after onset of aSAH symptoms (mRS 0-2/mRS 3-6).
Time Frame
at 3 months, at 1 year and at 2 years after onset of aSAH symptoms
Title
Activity of microglia cells assessed with PET
Description
It will be explored whether [11C](R)-PK11195 can be used to test the hypothesis of neuroprotective effect of xenon and to explore the role of inflammatory process for DCI after SAH. This could be demonstrated by showing less microglial activation in xenon group than in the reference therapy group and in the patients with good outcome, i.e. no DCI; Difference of activity of microglia cells between xenon and control group and in predicting risk for DCI
Time Frame
DCI between day 4 and 6 weeks after onset of aSAH symptoms; The 1st PETscan 4 ±1 weeks after onset of aSAH symptoms and the 2nd scan at 3 months after onset of SAH symptoms.
Title
Activity of microglia cells assessed with PET
Description
It will be explored whether [11C](R)-PK11195 can be used to test the hypothesis of neuroprotective effect of xenon and to explore the role of inflammatory process for neurological outcome after SAH. This could be demonstrated by showing less microglial activation in xenon group than in the reference therapy group and in the patients with good outcome, i.e. mRS 0-2;
Time Frame
The 1st scan at 4 ±1 weeks after and the 2nd scan at 3 months after onset of SAH symptoms. Outcome: at 3 months, at 1 year and at 2 years after onset of aSAH symptoms
Title
Cerebral fluid dynamics
Description
Predictive value of CFD simulations assessed with 3 dimensional DSA within 4 days of ICU arrival in predicting risk for EBI within 72 hours after onset of aSAH symptoms
Time Frame
Measures performed within 72 hours of ICU arrival
Title
Cerebral fluid dynamics
Description
Predictive value of CFD simulations assessed with 3 dimensional DSA within 21 days of ICU arrival in predicting risk for neurological outcome at 3 months, at 1 year and at 2 years after SAH (mRS 0-2)
Time Frame
Measures performed within 21 days of ICU arrival; outcome at 3 months, at 1 year and at 2 years after onset of aSAH symptoms
Title
Cerebral fluid dynamics
Description
Predictive value of CFD simulations assessed with 3 dimensional DSA within 21 days of ICU arrival in predicting risk for DCI within 6 weeks after onset of aSAH symptoms
Time Frame
Measures performed within 21 days of ICU arrival; DCI within 6 weeks after onset of aSAH symptoms

10. Eligibility

Sex
All
Minimum Age & Unit of Time
18 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
Inclusion Criteria: To be considered eligible to participate in this study, a SAH subject must meet the inclusion criteria listed below: Informed consent obtained from the next of kin or legal representative Aneurysmal subarachnoid hemorrhage visible on CTA or DSA. Deterioration of consciousness to Hunt-Hess 3-5 Age of ≥ 18 years Intubated. GCS 3-12 obtained off neuromuscular blocking agents Xenon treatment can be started within 6 hours after onset of SAH symptoms Exclusion Criteria: An aSAH subject may not be enrolled in the trial if he/she meets any one of the exclusion criteria below: Acute or chronic traumatic brain injury Maximum diameter of intracerebral hemorrhage > 2.5 cm Pneumothorax or pneumomediastinum, Acute lung injury requiring ≥ 60% FIO2 (fraction of inspired oxygen). Systolic arterial pressure < 80 mmHg or mean arterial pressure < 60 mmHg for over 30 min period Bilaterally fixed and dilated pupils Positive pregnancy test, known pregnancy, or current breast-feeding Neurological deficiency due to traumatic brain injury or other neurological illness Imminent death or current life-threatening disease Current enrollment in another interventional study The subject is known to have clinically significant laboratory abnormality, medical condition (such as decompensated liver disease or severe chronic obstructive pulmonary disease), or social circumstance that, in the investigator's opinion, makes it inappropriate for the subject to participate in this clinical trial. Presence of implants or foreign bodies which are not known to be MRI safe
Central Contact Person:
First Name & Middle Initial & Last Name or Official Title & Degree
Timo T Laitio, MD, PhD
Phone
+358504653201
Email
timo.laitio@tyks.fi
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Timo T Laitio, MD, PhD
Organizational Affiliation
Turku University Hospital and University of Turku, Turku , Finland
Official's Role
Principal Investigator
Facility Information:
Facility Name
Aalto University School of Science
City
Helsinki
Country
Finland
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Timo Roine, PhD
Email
timo.roine@gmail.com
Facility Name
Kuopio University Hospital
City
Kuopio
Country
Finland
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Stepani Bendel
Email
stepani.bendel@kuh.fi
Facility Name
Tampere University Hospital
City
Tampere
Country
Finland
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Sari Karlsson, MD, PhD
Email
sari.karlsson@pshp.fi
Facility Name
Turku University Hospital
City
Turku
ZIP/Postal Code
20521
Country
Finland
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Timo T Laitio, MD, PhD
Phone
+358504653201
Email
timo.laitio@tyks.fi
Facility Name
Elomatic
City
Turku
ZIP/Postal Code
20810
Country
Finland
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Juha Tanttari, MSc
Facility Name
University of Turku, Turku Bioscience, Analysis of the metabolomics
City
Turku
Country
Finland
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Matej Orešič, PhD
Email
matej.oresic@utu.fi
First Name & Middle Initial & Last Name & Degree
Alex Dickens, PhD
Email
alex.dickens@utu.fi
Facility Name
Örebro University
City
Örebro
Country
Sweden
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Tuulia Hyötyläinen, PhD

12. IPD Sharing Statement

Plan to Share IPD
Yes
IPD Sharing Plan Description
The data of this study will be available to investigators whose proposed use of the data has been approved by an independent review committee. Individual participant data that underlie the results reported in this Article will be shared (text, tables, figures, and appendices), after de-identification, along with the study protocol. These data will be available 6 months after the Article's pulication and will be available for 12 months from publication. Data can be used for individual participant data meta-analysis. Requests and proposals should be directed to timo.laitio@elisanet.fi. To gain access, data requestors will need to sign a data access agreement.
IPD Sharing Time Frame
data will be available 6 months after the Article's pulication and will be available for 12 months from publication.
IPD Sharing Access Criteria
Requests and proposals should be directed to timo.laitio@elisanet.fi. To gain access, data requestors will need to sign a data access agreement.
Citations:
PubMed Identifier
26978207
Citation
Laitio R, Hynninen M, Arola O, Virtanen S, Parkkola R, Saunavaara J, Roine RO, Gronlund J, Ylikoski E, Wennervirta J, Backlund M, Silvasti P, Nukarinen E, Tiainen M, Saraste A, Pietila M, Airaksinen J, Valanne L, Martola J, Silvennoinen H, Scheinin H, Harjola VP, Niiranen J, Korpi K, Varpula M, Inkinen O, Olkkola KT, Maze M, Vahlberg T, Laitio T. Effect of Inhaled Xenon on Cerebral White Matter Damage in Comatose Survivors of Out-of-Hospital Cardiac Arrest: A Randomized Clinical Trial. JAMA. 2016 Mar 15;315(11):1120-8. doi: 10.1001/jama.2016.1933.
Results Reference
result
PubMed Identifier
29169472
Citation
Arola O, Saraste A, Laitio R, Airaksinen J, Hynninen M, Backlund M, Ylikoski E, Wennervirta J, Pietila M, Roine RO, Harjola VP, Niiranen J, Korpi K, Varpula M, Scheinin H, Maze M, Vahlberg T, Laitio T; Xe-HYPOTHECA Study Group. Inhaled Xenon Attenuates Myocardial Damage in Comatose Survivors of Out-of-Hospital Cardiac Arrest: The Xe-Hypotheca Trial. J Am Coll Cardiol. 2017 Nov 28;70(21):2652-2660. doi: 10.1016/j.jacc.2017.09.1088.
Results Reference
result

Learn more about this trial

Effect of Xenon on Brain Injury After Aneurysmal Subarachnoid Hemorrhage

We'll reach out to this number within 24 hrs