Skip to main content
Erschienen in: Acta Neurochirurgica 6/2023

Open Access 27.04.2023 | Original Article

Safety and technical efficacy of early minimally invasive endoscopy-guided surgery for intracerebral haemorrhage: the Dutch Intracerebral haemorrhage Surgery Trial pilot study

verfasst von: Lotte Sondag, Floris H.B.M. Schreuder, Sjoert A.H. Pegge, Jonathan M. Coutinho, Diederik W.J. Dippel, Paula M. Janssen, W. Peter Vandertop, Hieronymus D. Boogaarts, Ruben Dammers, Catharina J.M. Klijn, On behalf of the Dutch ICH Surgery Trial Study Group, part of the CONTRAST consortium

Erschienen in: Acta Neurochirurgica | Ausgabe 6/2023

Abstract

Background

Previous randomised controlled trials could not demonstrate that surgical evacuation of intracerebral haemorrhage (ICH) improves functional outcome. Increasing evidence suggests that minimally invasive surgery may be beneficial, in particular when performed early after symptom onset. The aim of this study was to investigate safety and technical efficacy of early minimally invasive endoscopy-guided surgery in patients with spontaneous supratentorial ICH.

Methods

The Dutch Intracerebral Haemorrhage Surgery Trial pilot study was a prospective intervention study with blinded outcome assessment in three neurosurgical centres in the Netherlands. We included adult patients with spontaneous supratentorial ICH ≥10mL and National Institute of Health Stroke Scale (NIHSS) score ≥2 for minimally invasive endoscopy-guided surgery within 8 h after symptom onset in addition to medical management. Primary safety outcome was death or increase in NIHSS ≥4 points at 24 h. Secondary safety outcomes were procedure-related serious adverse events (SAEs) within 7 days and death within 30 days. Primary technical efficacy outcome was ICH volume reduction (%) at 24 h.

Results

We included 40 patients (median age 61 years; IQR 51–67; 28 men). Median baseline NIHSS was 19.5 (IQR 13.3–22.0) and median ICH volume 47.7mL (IQR 29.4–72.0). Six patients had a primary safety outcome, of whom two already deteriorated before surgery and one died within 24 h. Sixteen other SAEs were reported within 7 days in 11 patients (of whom two patients that already had a primary safety outcome), none device related. In total, four (10%) patients died within 30 days. Median ICH volume reduction at 24 h was 78% (IQR 50–89) and median postoperative ICH volume 10.5mL (IQR 5.1–23.8).

Conclusions

Minimally invasive endoscopy-guided surgery within 8 h after symptom onset for supratentorial ICH appears to be safe and can effectively reduce ICH volume. Randomised controlled trials are needed to determine whether this intervention also improves functional outcome.

Trial registration

Clinicaltrials.​gov: NCT03608423, August 1st, 2018.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s00701-023-05599-2.
Ruben Dammers and Catharina J.M. Klijn contributed equally.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CTA
Computed tomography angiography
DSMB
Data Safety Monitoring Board
EVD
External ventricular drain
GCS
Glasgow Coma Scale score
ICH
Intracerebral haemorrhage
ICH-GS
ICH-Grading Scale
IQR
Interquartile range
INR
International normalised ratio
IVH
Intraventricular haemorrhage
mRS
modified Rankin Scale
NIHSS
National Institute of Health Stroke Scale
NCCT
Non-contrast computed tomography
OR
Odds ratio
PHO
Perihematomal oedema
RCT
Randomised controlled trial
SAE
Serious adverse event
TIA
Transient ischemic attack

Introduction

Spontaneous intracerebral haemorrhage (ICH) accounts for 16–19% of all strokes in the Western world and for 28–32% in low- and middle-income countries [12, 24]. ICH contributes profoundly to stroke-related disability and 30-day case fatality is around 40% [38, 48]. Survival and functional outcome after spontaneous ICH are independently predicted by age, Glasgow Coma Scale (GCS) score, haematoma volume, the presence of intraventricular haemorrhage (IVH) [38], and hematoma growth [9, 40]. Around 20% of patients with ICH show growth of hematoma, mainly within the first 3 h after symptom onset [3]. Neurotoxicity, caused by blood degradation products and plasma-derived components such as thrombin, and inflammation, may lead to the development of perihematomal oedema (PHO). This process starts already within 3 to 4 h after symptom onset [4, 20]. Reduction of the hematoma volume in an early stage may not only alleviate the direct mass effect of the ICH, but may also prevent hematoma growth and development of PHO [35, 51]. Theoretically, this could result in better functional outcome [2, 35]. Increasing evidence including recent narrative [8, 14] and systematic reviews [43, 44] of randomised controlled trials (RCTs) suggests that surgical hematoma evacuation may reduce mortality and improve functional outcome [13, 43, 44, 55]. However, large high-quality RCTs have not established this presumed beneficial effect of surgery [15, 16, 30, 31, 44]. Besides evacuation by means of a craniotomy, several minimally invasive techniques are available to remove the hematoma [2, 17, 36, 41, 55]. Recent case series suggest that endoscopy-guided minimally invasive evacuation by means of aspiration is promising and safe and effectively reduces haemorrhage volume, with few complications [1, 11, 19, 23, 27, 41, 45, 46, 53]. The MISTIE III trial recently failed to demonstrate a beneficial effect on functional outcome of minimally invasive surgery with local application of alteplase followed by hematoma aspiration up to 72 h [16]. The relatively long median time from symptom onset to surgery in MISTIE III (59 h) [16] and in the other large, multicentre, high-quality studies of conventional surgery STICH (30 h) [30] and STICH-II (26 h) [31] might in part explain the lack of a treatment effect. An individual patient data meta-analysis of RCTs found that outcome improved after surgery compared to standard medical management, if surgery was performed within 8 h [13]. However, one study showed that ultra-early surgery (within 4 h) was associated with rebleeding [34], whilst other studies have shown that ultra-early surgery (<4–6 h) is safe [25, 26]. In addition, a recent study showed that for every hour that patients were operated earlier, there was a 5% increase in the odds of good functional outcome at 6 months [22].
Well-designed RCTs are needed to provide definitive and guideline changing evidence whether or not early minimally invasive and endoscopy-guided surgical evacuation would reduce mortality after sICH and improves functional outcome [43]. As a prelude to such a large randomised clinical trial, the aim of the Dutch ICH Surgery Trial (DIST) pilot study was to investigate safety and technical efficacy of minimally invasive endoscopy-guided surgery for the treatment of spontaneous supratentorial ICH within 8h after symptom onset.

Methods

Study design

The DIST pilot study was a multicentre, prospective, non-randomised intervention study in the Netherlands, with blinded endpoint assessment, investigating the safety and technical efficacy of minimally invasive endoscopy-guided surgical evacuation of supratentorial spontaneous ICH within 8h after symptom onset, in addition to standard medical management (Clinicaltrials.​gov: NCT03608423; http://​dutch-ich.​nl). Patients underwent minimally invasive endoscopy-guided surgery in addition to standard medical management in three hospitals. In seven other centres, patients received standard medical management only.
We aimed to include 40 patients in the surgical arm, with a minimum of 10 patients per centre, to be able to obtain clinically meaningful data on the safety and technical efficacy of our intervention, assuming we could show that on average we would be able to reduce ICH volume with at least 60%.
Here, we report the results of the 40 patients who underwent surgery. This study was approved by the medical ethics committee of the Erasmus MC University Medical Centre Rotterdam (MEC_2018-012).

Participants

Patients were eligible if they met the following inclusion criteria: age ≥18 years, supratentorial spontaneous (non-traumatic) ICH ≥10 mL confirmed by non-contrast computed tomography (NCCT) scan, National Institutes of Health Stroke Scale (NIHSS) score ≥2, and initiation of surgery possible within 8 h after symptom onset. Written informed consent was obtained from all patients or their legal representatives. The diagnosis of spontaneous ICH was based on the absence of a causative macrovascular lesion (i.e. aneurysm, arteriovenous malformation, dural arteriovenous fistula, cerebral venous sinus thrombosis) on computed tomography angiography (CTA) scan performed immediately following the NCCT [18, 49].
ICH volumes in the acute setting were calculated using the ABC/2 method. We verified these volumes if they were close to the limit of 10mL using volume measurements in Brainlab® (AG, München, Germany). Screening logs were not kept. Patients were excluded from participation if they had any other known cause of the ICH (i.e. tumour, cavernoma), pre-stroke disability that interfered with the assessment of functional outcome (i.e. pre-stroke modified Rankin Scale (mRS) score >2) [6], untreated coagulation abnormalities, pregnancy, or when a patient was moribund (i.e. coning, dilated unresponsive pupils). Patients who were using a vitamin K antagonist could be included after reversal of the anticoagulation effect (INR ≤1.3). Patients who were using direct oral anticoagulants were excluded. The use of antiplatelet therapy, including clopidogrel, was not an exclusion criterion.
At baseline, we collected data on age and sex, medical history (presence of hypertension, atrial fibrillation, diabetes mellitus, hypercholesterolemia, previous ischemic or haemorrhagic stroke, transient ischemic attack, previous major bleeding, myocardial infarction, premorbid cognitive dysfunction, premorbid disability on the mRS), and the use of medication (antiplatelet therapy, antihypertensive agents, oral anticoagulants, statins). In addition, we collected systolic and diastolic blood pressure, GCS score, NIHSS score, and ICH-Grading Scale (ICH-GS, with higher scores indicating worse prognosis) score [42] at baseline. An independent neuroradiologist assessed baseline NCCTs for ICH volume, ICH location, presence of intraventricular, subdural or subarachnoid extension, PHO volume, hydrocephalus, and CTA for presence of a spot sign, blinded to clinical characteristics and to safety and technical efficacy outcomes. The same independent and blinded neuroradiologist assessed the NCCT that was performed at 24 h. We calculated time intervals from symptom onset (or last seen well) to NCCT. All patients underwent assessment of the NIHSS at 24 h and 7 days or discharge. All SAEs within 30 days were reported.

Intervention

Neurosurgeons with experience in endoscopic neurosurgery for other purposes were trained to perform the surgical procedures. They followed a prespecified protocol (Online resource data) and performed surgery under general anaesthesia. The baseline NCCT was uploaded into neuronavigational software (Brainlab® AG, München, Germany). Neurosurgeons selected a trajectory for evacuation that allowed safe access to the longest possible axis of the haemorrhage, in a similar way as in the ICES study [50], and described earlier as the stereotactic intracerebral haemorrhage underwater blood aspiration (SCUBA) and ADAPT technique [21, 41]. The image-guidance probe was positioned over the candidate entry point to assess whether or not the endoscope sheath would transgress any critical functional areas. Neurosurgeons created a burr hole of 1.5–2.0 cm, opened the dura, and incised and coagulated the cortical surface. A localisation array was attached to the selected neuro-endoscopic sheath and registered to the navigation system. The sheath was stabilised into the target zone, typically at 2/3 of the length of the long axis, using neuronavigation. The neuro-endoscope was inserted into the sheath and under direct visualisation the ArtemisTM Neuro Evacuation Device (Penumbra Inc. Alameda, California, USA) was placed through the working channel of the endoscope. Intermittent irrigation through the endoscope and hematoma aspiration via the ArtemisTM device was performed under direct visualisation until a clear working view was created, revealing the hematoma cavity. This was repeated until no further clot could be evacuated at this location. In case of active bleeding, irrigation was continued until the bleeding stopped. Once haemostasis was obtained, the endoscope sheath was retracted to a point approximately 1/3 of the hematoma cavity. The suctioning and irrigation process was then repeated at this point. Suctioning was continued until the surgeon estimated to have removed at least 75–80% of the hematoma volume. Then, the endoscope was introduced for a final inspection of the cavity to check for any ongoing bleeding that required additional treatment through irrigation and occasional bipolar coagulation. The dura and skin were closed. An NCCT was performed intra-operatively or immediately after evacuation to confirm adequate haemorrhage evacuation. Whether or not to opt for resumption of the surgery to evacuate any residual haemorrhage was at the surgeons’ discretion. Patients were admitted to the neuro-intensive care unit or a dedicated stroke unit, and were treated according to the Dutch national stroke guideline [10]. We recorded time from symptom onset to arrival in the operating room, to first incision and to skin closure, blood pressure measurements during surgery, the type of endoscope that was used, the occurrence of active bleeding or rebleeding during surgery, conversion to craniotomy, and whether the surgery was performed in a hybrid operating room.

Outcomes

The primary safety outcome was death within 24 h or neurological deterioration with an increase of ≥4 points on the NIHSS at 24 h. Primary safety outcomes were adjudicated by an independent committee, consisting of two neurologists and one neurosurgeon. Secondary safety outcomes were ICH volume at 24 h, death at 7 and 30 days, and 7-day procedure-related complications. Additionally, we recorded all SAEs within 30 days. The primary technical efficacy outcome was the percentage of volume reduction comparing the ICH volumes on the NCCT at baseline and the NCCT at 24 h. All images were centrally assessed by an independent neuroradiologist, blinded for baseline characteristics and safety and technical efficacy outcomes. Secondary technical efficacy outcomes were the percentage of patients with a clot volume reduction of ≥60%, the percentage of patients with a clot volume reduction ≥80%, the percentage of patients with a remaining clot volume ≤15mL at 24 h, and the number of patients in whom the minimally invasive procedure was converted to craniotomy. (Re)bleed was defined as a higher ICH volume at 24 h than at baseline or postoperatively (irrespective of change in clinical symptoms). The Data Safety Monitoring Board (DSMB) would consider to advise to hold the study when the lower limit of the 95% CI of the proportion of patients with the occurrence of the primary safety outcome reached 40%.

Data analyses

Baseline characteristics are displayed as number of patients with corresponding percentages, mean with standard deviation (SD), or median with 25% and 75% interquartile range (IQR) as appropriate. ICH volume at baseline and 24 h NCCT, and IVH volume and PHO volume at baseline NCCT, were measured via manual segmentation using ITK-SNAP 3.8 (http://​www.​itksnap.​org) [54] and volumes were calculated using MATLAB 2018a, based on the number of voxels and voxel size in three directions. For safety outcomes, we calculated absolute numbers and percentages with 95% confidence intervals (CIs). An independent DSMB monitored the study with the aim to advise to stop the study if the lower limit of the 95% CI of the percentage of patients with a primary safety outcome would reach 40%. Reduction of hematoma volume was expressed as median (IQR) percentage volume reduction. To obtain insight into the effect of a potential learning curve on technical efficacy, we assessed the first and second half of patients separately as an exploratory analysis. We split the group by the first half and second half within each centre. In case of unequal numbers, the second group would be the largest.

Results

Forty patients were included between March 2019 and January 2021. We experienced delays at the start due to limited availability of the surgical devices, and during the study because of the COVID-19 pandemic with temporary halting of clinical research activities and limited availability of operation room capacity and ICU beds. Demographics and baseline characteristics are summarised in Table 1. Median ICH volume at baseline was 47.7mL (IQR 29.4–72.0mL; 2 patients had a baseline ICH volume <15mL). Median time from symptom onset to the start of the surgery (first incision) was 6 h and 43 min (IQR 5 h 26 min–7 h 56 min). Median duration of the procedure (first incision to skin closure) was 68 min (IQR 55–106 min). Median ICH volume after 24 h was 10.5mL (IQR 5.1–23.8mL). Further details of the surgical procedures are summarised in Table 2. Online resource table 2 shows the baseline characteristics and technical outcome parameters according to baseline ICH volume subgroups (<30mL and ≥30mL). An illustrative case is shown in Fig. 1. Twelve patients had a CTA spot sign, of whom five had active bleeding during surgery. Of a total of 19 patients with active bleeding during surgery (Table 2), 14 did not have a spot sign at baseline.
Table 1
Patient characteristics
Characteristic
N=40
Age, mean (SD)
59.5 (13.6)
Men, n (%)
28 (70.0)
Medical history of
 
 Hypertension, n (%)
14 (35.0)
 Atrial fibrillation, n (%)
4 (10.0)
 Diabetes mellitus, n (%)
6 (15.0)
 Hypercholesterolemia, n (%)
9 (22.5)
 Ischemic stroke, n (%)
1 (2.5)
 TIA, n (%)
5 (12.5)
 Previous ICH, n (%)
2 (5.0)
 Myocardial infarction, n (%)
3 (7.5)
 Premorbid cognitive dysfunction, n (%)
1 (2.5)
Medication at baseline, use of
 
 Antiplatelet therapy, n (%)
11 (27.5)
  Acetylsalicylic acid
7 (17.5)
  Clopidogrel
4 (10.0)
 Antihypertensive drug(s), n (%)
11 (27.5)
 Vitamin K antagonist, n (%)
1 (2.5)
Statin, n (%)
13 (32.5)
Pre-ICH mRS score 0, n (%)
33 (82.5)
Pre-ICH mRS score 1, n (%)
5 (12.5)
Pre-ICH mRS score 2, n (%)
2 (5.0)
Time from symptom onset or last seen well to arrival study centre, median (IQR)
3h 32min (1h 49min–4h 52min)
Glasgow Coma Scale score (on admission), median (IQR)
12 (9–14)
 GCS score <9, n (%)
5 (12.5)
 GCS score 9–12, n (%)
17 (42.5)
 GCS score 13–15, n (%)
18 (45)
NIHSS score (on admission), median (IQR)
19 (13–22)
Systolic blood pressure, mmHg (on admission), mean (SD)
174 (31)
Diastolic blood pressure, mmHg (on admission), mean (SD)
102 (28)
Baseline NCCT scan
 
 ICH volume (mL), mean (SD)
50 (23)
 Lobar location, n (%)
11 (27.5)
 Left hemispheric location, n (%)
21 (52.5)
 IVH present, n (%)
19 (47.5)
 IVH volume (mL), median (IQR)
0 (0-17)
 Hydrocephalus, n (%)
11 (27.5)
 Perihematomal oedema volume (mL), median (IQR)a
14 (7-29)
 Subarachnoid extension of ICH, n (%)
9 (22.5)
 Finger like projections, n (%)
9 (22.5)
 Time from onset symptoms (or last seen well) to baseline NCCT, median (IQR)
3h 46min (1h 30min–5h 33min)
CT-angiography spot sign present, n (%)b
12 (30)
ICH-GS score at inclusion, n (%)
 
 ICH-GS-score 5
1 (2.5)
 ICH-GS-score 6
3 (7.5)
 ICH-GS-score 7
9 (22.5)
 ICH-GS-score 8
10 (25.0)
 ICH-GS-score 9
11 (27.5)
 ICH-GS-score 10
4 (10.0)
 ICH-GS-score 11
2 (5.0)
CT computed tomography scan, GCS Glasgow Coma Scale, ICH intracerebral haemorrhage, ICH-GS intracerebral haemorrhage grading scale, IQR interquartile range, IVH intraventricular haemorrhage (extension), mRS modified Rankin Scale, NIHSS National Institutes of Health Stroke Scale, TIA transient ischemic attack
aTwo patients had no perihematomal oedema
bNo CTA available in 2 patients
Table 2
Surgical treatment
 
N=40
Time from symptom onset to arrival operating room (hours, min), median (IQR)
5h 53 (4h 15–7h 14)
 Interval from symptom onset or last seen well to first cut <4 hours, n (%)
3 (7.5)
 Interval from symptom onset or last seen well to first cut <6 hours, n (%)
21 (52.5)
Surgery performed on hybrid OR, n (%)
8 (20.0)
Endoscope used, n (%)
 
 Lotta (Storz)
39 (97.5)
 Minop (B.Braun)
1 (2.5)
Active bleeding during surgery, n (%)
19 (47.5)
Rebleeding or new intracranial bleeding during surgery, n (%)
3 (7.5)
Highest systolic blood pressure during surgery (mmHg), median (IQR)
162 (139–189)
Lowest systolic blood pressure during surgery (mmHg), median (IQR)
117 (97–140)
Re-operation after intra-operative or direct post-operative NCCT, n (%)a
8 (20.0)
EVD placement, n (%)b
9 (22%)
IQR interquartile range, OR operation room
aIn these patients, median duration of surgery (from first cut to skin closure after resumption of operation) was 2 h and 58 min
bEVD was placed either during the initial surgical procedure, or in a later stadium during admission

Primary safety outcomes

Of the 40 included patients, six (15%; 95% CI 5.7–29.9%) had a primary safety outcome. One patient died within 24 h (2.5%; 95% CI: 0.1–13.2%) and five patients (12.5%; 95% CI: 4.2–26.8%) experienced an increase in NIHSS score of ≥4 points at 24 h, compared to baseline. Two of these five patients deteriorated in the emergency room before surgery had commenced. Two other patients of these five patients remained intubated at 24 h (but not at baseline), which increased their NIHSS scores. The fifth patient had disorientation in time and an increase in the severity of a left-sided paresis at 24 h, which partly recovered over time. Further clinical details of the primary outcomes are outlined in Online resource Table 3.

Technical efficacy

The median percentage volume reduction on CT at 24 h was 78% (IQR 50–89%) compared to baseline NCCT. Table 3 summarises the secondary safety and technical efficacy outcomes. In the majority of patients, the ICH volume was reduced with 60% or more and the remaining volume ≤15mL. In one patient (2.5%), the surgical procedure was converted to a craniotomy because of continuing active bleeding. Median proportional ICH volume reduction in the first half of patients was 70.5% (IQR 35.5–86.2) and 82.6% (IQR 55.4–89.8) in the second half of patients, whilst case mix appeared largely comparable (Table 4; Online resource Fig).
Table 3
Secondary safety and efficacy outcomes
 
N = 40
ICH volume at 24 h (mL), median (IQR)
10.5 (5.1–23.8)
Secondary safety outcomes, n (%)
 
 Death within 7 days
1 (2.5%)
 Death within 30 days
4 (10.0%)
 Serious adverse events <7 days, excluding primary safety outcomes
15 (in 11 patients)
 Serious adverse events between 8 and 30 days
22 (in 13 patients)
Secondary technical efficacy outcomes, n (%)
 
 Patients with clot volume reduction ≥60%
28 (70.0)
 Patients with clot volume reduction ≥80%
19 (47.5)
 Patients with remaining clot volume ≤15mL
23 (57.5)
 Conversion to craniotomy
1 (2.5)
ICH intracerebral haemorrhage, IQR interquartile range
Table 4
Baseline characteristics and surgical results in first and second half of surgical patients
Baseline characteristics
First half of surgical patients ( n=19)
Second half of surgical patients (n=21)
Age (years), median (IQR)
57 (45–63)
63 (57.5–69.5)
Men, n (%)
13 (68)
15 (71)
ICH volume baseline (mL), median (IQR)
49.7 (29.4–73.7)
42.9 (28.9–65.9)
IVH, n (%)
11 (57.9)
8 (38.1)
NIHSS baseline, median (IQR)
20 (19–22)
17 (9.5–20.5)
GCS baseline, median (IQR)
10 (9–13)
13 (10–14.5)
ICH-GS score, median (IQR)
9 (7–9)
8 (7–9)
Outcomes
  
Time from symptom onset or last seen well to first incision, median (IQR)
6h 35min (5h 19min–8h 10min)
7h 12min (5h 35min–7h 55min)
Postoperative remaining volume (mL), median (IQR)
16.7 (5.0–30.9)
8.4 (4.4–23.0)
Absolute ICH volume reduction (mL), median (IQR)
26.6 (15.5–41.6)
35.9 (19.3–50.1)
ICH volume reduction ≥80%, n (%)
6 (31.6)
13 (61.9)
ICH volume reduction ≥60%, n (%)
12 (63.2)
16 (76.2)
Postoperative remaining volume ≤15mL
9 (47.4)
14 (66.7)
Duration of the procedure (first incision to skin closure) median min (IQR)
84.0 (52.0–112.0)
62.0 (55.5–89.5)
GCS Glasgow Coma Scale score, ICH intracerebral haemorrhage, ICH-GS intracerebral haemorrhage grading scale, IQR interquartile range, IVH intraventricular haemorrhage (extension), NIHSS National Institutes of Health Stroke Scale

Secondary safety outcomes

Case fatality at 30 days was 10% (95% CI 2.5–23.7%, n=4). Besides the primary safety outcomes, 16 SAEs within the first 7 days were reported in 11 patients, of whom 2 patients that also had a primary safety outcome (Table 5). Another 23 SAEs were registered between 8 and 30 days in 13 patients. Of these 13 patients, 11 patients also had an earlier SAE reported within 7 days, of whom two also had a primary safety outcome. No device-related SAEs were reported. One patient had an intracranial infection within 7 days (at day 4) with a positive cerebrospinal fluid (CSF) culture. None of the 9 patients that were treated with an external ventricular drain (EVD) developed an intracranial infection proven with positive CSF cultures. Three of these patients (7.5%) were pragmatically treated for a suspected intracranial infection, based on clinical symptoms (n=2) or increased leukocyte count in the CSF without a decreased glucose level (n=1) in addition to clinical symptoms, but CSF cultures were not performed or remained negative.
Table 5
Overview of SAEs (primary safety outcomes excluded) per organ system
 
SAE ≤7 days
SAE 8–30 days
Gastro-intestinal disorders
2
Infections and infestations (other than intracranial infection or pneumonia)
1
1
Nervous system disorders
  
 ICH progression (CT confirmed)
1a
2c
 Clinical deterioration; ICH progression suspected but not CT confirmed
2b
1
 ICH progression with epidural hematoma
1c
 ICH progression with hydrocephalus
1
 Intracranial infection, with positive CSF culture
1d,e
 Suspected intracranial infection (without positive CSF culture)
4d,e
 Postoperative site infection
1d
 Other
1
Psychiatric disorders
3
1
Renal and urinary disorders
1
1
Respiratory, thoracic and mediastinal disorders
  
 Pneumonia
5
3
 Pulmonary embolism
0
1
Reintubation
2
2
Other
0
1
Total
16
23
ICH intracerebral haemorrhage
aOne patient had ICH progression at the day of surgery with less than four points NIHSS increase
bTwo patients deteriorated clinically but no imaging was performed to confirm ICH progression
cAll three occurred in the same patient
dOne patient had an intracranial infection within seven days as well as a postoperative site infection followed by an intracranial infection between eight and 30 days
eOne patient had an intracranial infection on day four, with a positive cerebrospinal fluid (CSF) culture for Streptococcus mitis. This patient was not treated with an EVD. Of the other three patients that had a suspected intracranial infection after 7 days, all have had an EVD and none of them had positive CSF cultures
Four patients (10%) had a rebleed within 30 days, of whom two within 7 days. In two patients, the rebleed occurred on the first day (3.5 and 3 h after surgery that was started 4 h 15 min and 8 h 13 min after symptom onset). In the other two patients (surgery started after 7 h 40 min and 4 h 11 min after symptom onset), a rebleeding occurred at day 8 and 11. In the latter, therapeutic heparin was started because of a pulmonary embolism. All four patients with a rebleed underwent another surgical procedure: in two an EVD was placed, one underwent a hemicraniectomy with hematoma evacuation and another patient was treated with bilateral EVD-placement and craniotomy. In this 63-year-old patient with a 48-mL lobar hematoma, a diagnosis of cerebral amyloid angiopathy was suspected. In one of the patients treated with EVD, uncontrolled postoperative hypertension may have contributed to the ICH progression. A full list of SAEs per organ system can be found in Table 5.

Discussion

In the DIST pilot study, minimally invasive endoscopy-guided surgery within 8 h of symptom onset in patients with a spontaneous supratentorial ICH appeared safe and could be performed with good technical results. Technical efficacy tended to become better in the second half of the study, suggesting a learning curve for the procedure.
Our study differs from previous studies in that all patients had minimally invasive endoscopy-guided hematoma evacuation within the time window of 8 h after symptom onset. Previous non-randomised studies of endoscopy-guided minimally invasive surgery included patients up to 16 days after symptom onset with a median time to surgery of 19 h to 2.3 days [23, 46, 50]. In these studies, only a minority of patients was operated on within 8 h after symptom onset. The optimal timing of surgical evacuation is still a topic for debate. In a systematic review and meta-regression analysis, we recently showed that surgery seemed to be more effective when performed earlier after symptom onset [44]. Another review of 15 high-quality studies including 2152 patients found no difference in the risk of death or dependence between minimally invasive surgery performed within 24 h (OR 0.49, 95% CI 0.38–0.63, five studies) or within 72 h (OR 0.57, 95% CI 0.43–0.76, 12 studies) as compared with medical care or conventional craniotomy [43]. In both reviews, analyses were based on study level data and not on individual patient data. In addition, only one of the included studies restricted inclusion to patients in whom surgery could be started within 8 h after symptom onset [37]. In an observational study in 59 patients without a spot sign on CTA, the authors found no difference in rebleeding rates after stereotactic aspiration within 6 h or after 6 h after symptom onset [26]. In contrast, a small pilot study that assessed hematoma evacuation by means of a craniotomy within 4 h after symptom onset was stopped early because rebleeding occurred in four of 11 patients within 24 h after surgery [34]. In our study, a rebleed did not occur in the three patients in whom surgery was started within 4 h after symptom onset, and occurred in only one of 21 patients in whom surgery was started within 6 h. In MISTIE III (median time between symptom onset and start of surgery 58 h), six of 255 patients (2%) had a symptomatic bleeding within 72 h, and 81 patients (32%) an asymptomatic bleeding [16]. In the ICES study [50] (median time between symptom onset and start of surgery 30 h), three of 14 patients (21%) had an asymptomatic bleeding within 72 h, defined as an increase of at least 5mL and less than 2 points increase on the GCS motor score. Although a previous cohort study in 143 patients with spontaneous ICH who underwent endoscopic hematoma evacuation found no association between hypertension and postoperative rebleeding [32], we cannot exclude that elevated blood pressure after surgery may have contributed to rebleeding in one of the patients in our study. In practice, we strive for a systolic blood pressure of <160 mmHg. We could not confirm the findings of previous studies that the CTA spot sign is associated with postoperative rebleeding [32, 33]. In addition, we found no association between the presence of a CTA spot sign and intra-operative active bleeding, in contrast to what has previously been suggested by others [33]. In that study, 23% of patients were operated within 8 h, and could also have infratentorial ICH, and more frequently had a coagulopathy [33] than in our study. Based on our results, we suggest that patients with a CTA spot sign should not be excluded from future trials. Our 30-day case fatality of 10% is similar to that of 9–14% found in MISTIE III [16], as well as in ICES [50] (7%, 95% CI 1.8–33.9%) and other prospective studies investigating minimally invasive endoscopy-guided surgery [23, 46], in which patients were operated on at a median of 30 h to 3.2 days after symptom onset [16, 23, 46, 50].
The low percentage of intracranial infection (n=1; 2.5%) in this study was similar to that in MISTIE III (n=2/255; 1%) [16], ICES (n=0/14) [50], and another prospective study investigating minimally invasive endoscopy-guided surgery (n=2/49; 4%) [53].
The technical efficacy in our study was similar to the 54 to 97% postoperative ICH volume reduction reported in other studies of various minimally invasive techniques performed at later times after symptom onset (median time between symptom onset to surgery ranging from 19 h to 2.3 days) [23, 28, 46, 47, 50]. Hence, technical efficacy appears no reason to defer surgery to later time-points.
Previous studies on endoscopic surgery have shown that the evacuation rate improves with more experience [5, 28, 50], and that better technical results are associated with improved functional outcomes [16]. In our study, there also appeared to be a learning curve, with a suggestion of slightly better technical results in the second part of the study (albeit with overlapping 95% confidence intervals). In MISTIE -III, a threshold of four patients per surgeon and seven per centre was found, after which no patient had poor end of treatment volumes [16]. This learning curve is important to take into account in the design of future studies. The IDEAL recommendations for the design and reporting of studies on surgical and interventional therapy innovations [29] advise to monitor the quality of the intervention, including pre-operative care, surgery, and post-operative care. For RCTs these recommendations additionally advise to evaluate the learning curve, ideally with Bayesian hierarchical models, and to minimise potential harms by mentoring and training, and to demonstrate that the technique can be widely adopted by surgeons [7, 29]. Because of the small sample size, we refrained from formal analysis of the learning curve.
Based on the results of this study and those of others, we conclude that high-quality RCTs are needed to investigate whether minimally invasive surgery improves functional outcome in patients with spontaneous supratentorial ICH. It remains to be determined whether the timing is indeed a key factor in the success of this treatment [44, 52], but ‘time is brain’ may also hold true for ICH [21, 39]. The existence of a learning curve should be taken into account in the design of such trials.
Several RCTs comparing minimally invasive endoscopy-guided surgery with standard medical management in different time windows are ongoing: MIND (NCT03342664) investigates minimally invasive surgery with the ArtemisTM device within 72 h after symptom onset, ENRICH (NCT02880878) investigates minimally invasive surgery with the Brainpath® device by NICO Corporation (Indianapolis, Indiana, USA) within 24 h after symptom onset, and EVACUATE (NCT04434807) is investigating minimally invasive aspiration by the Aurora Surgiscope® by Rebound Therapeutics® (Irvine, California, USA) within 8 h after symptom onset. In the Dutch Intracerebral Haemorrhage Surgery Trial (DIST, NCT05460793), we will investigate whether treatment with minimally invasive endoscopy-guided surgery within 8 hours after symptom onset in addition to medical management improves functional outcome after 6 months in comparison with medical management alone. DIST has started in October 2022 and is estimated to be completed in 2027. Strengths of the DIST pilot study are its prospective, multicentre design and the broad inclusion criteria, without limitations for age, GCS, or the presence of a CTA spot sign, following a standardised surgical protocol. In addition, all imaging to determine technical efficacy was centrally assessed by an independent neuroradiologist, blinded for baseline characteristics and outcomes. Furthermore, we assessed the difference in technical efficacy between the first half and second half of patients, highlighting the importance of training.
Our study also has some limitations. First, the sample size was small. Second, we were not able to report on the effect of early minimally invasive endoscopy-guided surgery on functional outcome. Third, we did not keep screening logs. Furthermore, results might not be generalisable to other countries than the Netherlands. In particular, the distance to a neurosurgical centre in the Netherlands is small in comparison with other countries.

Conclusion

This study shows that in patients with supratentorial ICH, minimally invasive endoscopy-guided surgery within 8 h after symptom onset appears safe and technically effective. An RCT is needed to assess whether this intervention improves functional outcome.

Acknowledgements

The Dutch Intracerebral Haemorrhage Surgery Trial pilot study—study group consists of the following: Lotte Sondag MD, Floris H.B.M. Schreuder MD PhD, Jelis Boiten MD PhD, Paul J.A.M. Brouwers MD PhD, Jonathan Coutinho MD PhD, Diederik W.J. Dippel MD PhD, M. Heleen den Hertog MD PhD, Paula M. Janssen MD, Wilmar M.T. Jolink MD PhD, L. Jaap Kappelle MD PhD, Kuan H. Kho MD PhD, Radboud W. Koot MD PhD, Paul L.M. de Kort MD PhD, Wouter A. Moojen MD PhD, Dharmin Nanda MD PhD, Onno P.M. Teernstra MD PhD, Bram van der Pol MD PhD, Inger R. de Ridder MD PhD, Marieke J.H. Wermer MD PhD, Albert van der Zwan MD PhD, W. Peter Vandertop MD PhD, Hieronymus D. Boogaarts MD PhD, Ruben Dammers MD PhD, Catharina J.M. Klijn MD PhD, Dana Holl, Anil Can. Study group authors’ affiliations can be found in Online resource Table 1. We would like to thank the Data Safety Monitoring Board, that consisted of the following: Craig Anderson (Chair), MD, PhD, FRACP, Director, The George Institute for Global Health and Professor of Neurology and Epidemiology, Faculty of Medicine, UNSW, Sydney, Australia, Ale Algra, MD, PhD, Professor of Clinical Epidemiology of Cerebrovascular Diseases, Utrecht Stroke Center, Department of Neurology and Neurosurgery and Julius Center University Medical Center Utrecht, The Netherlands. Wilco C. Peul, MD, PhD, MBA, Professor and Chair of Neurosurgery, Leiden University Department of Neurosurgery, The Hague & Leiden, the Netherlands.

Declarations

Ethical approval

All procedures performed were in accordance with the ethical standards of the institutional research committees and with the 1964 Helsinki Declaration and its later amendments. The study was approved by the medical ethics committee of the Erasmus MC University Medical Centre Rotterdam (MEC_2018-012).
Written informed consent was obtained from all patients or their legal representatives to participate in the study and to publish their data.

Conflict of interest

The authors declare no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

Die Chirurgie

Print-Titel

Das Abo mit mehr Tiefe

Mit der Zeitschrift Die Chirurgie erhalten Sie zusätzlich Online-Zugriff auf weitere 43 chirurgische Fachzeitschriften, CME-Fortbildungen, Webinare, Vorbereitungskursen zur Facharztprüfung und die digitale Enzyklopädie e.Medpedia.

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Anhänge

Supplementary information

Literatur
1.
Zurück zum Zitat Ali M, Zhang X, Ascanio LC et al (2022) Long-term functional independence after minimally invasive endoscopic intracerebral hemorrhage evacuation. J Neurosurg:1–11 Ali M, Zhang X, Ascanio LC et al (2022) Long-term functional independence after minimally invasive endoscopic intracerebral hemorrhage evacuation. J Neurosurg:1–11
2.
Zurück zum Zitat Al-Kawaz MN, Hanley DF, Ziai W (2020) Advances in therapeutic approaches for spontaneous intracerebral hemorrhage. Neurotherapeutics. Al-Kawaz MN, Hanley DF, Ziai W (2020) Advances in therapeutic approaches for spontaneous intracerebral hemorrhage. Neurotherapeutics.
3.
Zurück zum Zitat Al-Shahi Salman R, Frantzias J, Lee RJ et al (2018) Absolute risk and predictors of the growth of acute spontaneous intracerebral haemorrhage: a systematic review and meta-analysis of individual patient data. Lancet Neurol 17(10):885–894PubMedPubMedCentralCrossRef Al-Shahi Salman R, Frantzias J, Lee RJ et al (2018) Absolute risk and predictors of the growth of acute spontaneous intracerebral haemorrhage: a systematic review and meta-analysis of individual patient data. Lancet Neurol 17(10):885–894PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Askenase MH, Sansing LH (2016) Stages of the inflammatory response in pathology and tissue repair after intracerebral hemorrhage. Semin Neurol 36(3):288–297PubMedPubMedCentralCrossRef Askenase MH, Sansing LH (2016) Stages of the inflammatory response in pathology and tissue repair after intracerebral hemorrhage. Semin Neurol 36(3):288–297PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Awad IA, Polster SP, Carrion-Penagos J et al (2019) Surgical performance determines functional outcome benefit in the minimally invasive surgery plus recombinant tissue plasminogen activator for intracerebral hemorrhage evacuation (MISTIE) procedure. Neurosurgery 84(6):1157–1168PubMedPubMedCentralCrossRef Awad IA, Polster SP, Carrion-Penagos J et al (2019) Surgical performance determines functional outcome benefit in the minimally invasive surgery plus recombinant tissue plasminogen activator for intracerebral hemorrhage evacuation (MISTIE) procedure. Neurosurgery 84(6):1157–1168PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Bamford JM, Sandercock PA, Warlow CP, Slattery J (1989) Interobserver agreement for the assessment of handicap in stroke patients. Stroke. 20(6):828PubMedCrossRef Bamford JM, Sandercock PA, Warlow CP, Slattery J (1989) Interobserver agreement for the assessment of handicap in stroke patients. Stroke. 20(6):828PubMedCrossRef
7.
Zurück zum Zitat Cook JA, Ramsay CR, Fayers P (2004) Statistical evaluation of learning curve effects in surgical trials. Clin Trials 1(5):421–427PubMedCrossRef Cook JA, Ramsay CR, Fayers P (2004) Statistical evaluation of learning curve effects in surgical trials. Clin Trials 1(5):421–427PubMedCrossRef
8.
Zurück zum Zitat Cordonnier C, Demchuk A, Ziai W, Anderson CS (2018) Intracerebral haemorrhage: current approaches to acute management. Lancet. 392(10154):1257–1268PubMedCrossRef Cordonnier C, Demchuk A, Ziai W, Anderson CS (2018) Intracerebral haemorrhage: current approaches to acute management. Lancet. 392(10154):1257–1268PubMedCrossRef
9.
Zurück zum Zitat Davis SM, Broderick J, Hennerici M et al (2006) Hematoma growth is a determinant of mortality and poor outcome after intracerebral hemorrhage. Neurology. 66(8):1175–1181PubMedCrossRef Davis SM, Broderick J, Hennerici M et al (2006) Hematoma growth is a determinant of mortality and poor outcome after intracerebral hemorrhage. Neurology. 66(8):1175–1181PubMedCrossRef
10.
Zurück zum Zitat Dippel DWJ, van der Worp HB, Hofmeijer J et al (2021) Richtlijn herseninfarct en hersenbloedingen. Available at: https://richtlijnendatabasenl/richtlijn/herseninfarct_en_hersenbloeding/startpagina_herseninfarct_-bloedinghtml . Accessed 24 Sept 2017 Dippel DWJ, van der Worp HB, Hofmeijer J et al (2021) Richtlijn herseninfarct en hersenbloedingen. Available at: https://​richtlijnendatab​asenl/​richtlijn/​herseninfarct_​en_​hersenbloeding/​startpagina_​herseninfarct_​-bloedinghtml . Accessed 24 Sept 2017
11.
Zurück zum Zitat Fiorella D, Arthur A, Schafer S (2015) Minimally invasive cone beam CT-guided evacuation of parenchymal and ventricular hemorrhage using the Apollo system: proof of concept in a cadaver model. J Neurointerv Surg 7(8):569–573PubMedCrossRef Fiorella D, Arthur A, Schafer S (2015) Minimally invasive cone beam CT-guided evacuation of parenchymal and ventricular hemorrhage using the Apollo system: proof of concept in a cadaver model. J Neurointerv Surg 7(8):569–573PubMedCrossRef
12.
Zurück zum Zitat GBD-2019-Stroke-Collaborators (2021) Global, regional, and national burden of stroke and its risk factors, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Neurol 20(10):795–820CrossRef GBD-2019-Stroke-Collaborators (2021) Global, regional, and national burden of stroke and its risk factors, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Neurol 20(10):795–820CrossRef
13.
Zurück zum Zitat Gregson BA, Broderick JP, Auer LM et al (2012) Individual patient data subgroup meta-analysis of surgery for spontaneous supratentorial intracerebral hemorrhage. Stroke. 43(6):1496–1504PubMedPubMedCentralCrossRef Gregson BA, Broderick JP, Auer LM et al (2012) Individual patient data subgroup meta-analysis of surgery for spontaneous supratentorial intracerebral hemorrhage. Stroke. 43(6):1496–1504PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Gross BA, Jankowitz BT, Friedlander RM (2019) Cerebral intraparenchymal hemorrhage: a review. JAMA. 321(13):1295–1303PubMedCrossRef Gross BA, Jankowitz BT, Friedlander RM (2019) Cerebral intraparenchymal hemorrhage: a review. JAMA. 321(13):1295–1303PubMedCrossRef
15.
Zurück zum Zitat Hanley DF, Thompson RE, Muschelli J et al (2016) Safety and efficacy of minimally invasive surgery plus alteplase in intracerebral haemorrhage evacuation (MISTIE): a randomised, controlled, open-label, phase 2 trial. Lancet Neurol 15(12):1228–1237PubMedPubMedCentralCrossRef Hanley DF, Thompson RE, Muschelli J et al (2016) Safety and efficacy of minimally invasive surgery plus alteplase in intracerebral haemorrhage evacuation (MISTIE): a randomised, controlled, open-label, phase 2 trial. Lancet Neurol 15(12):1228–1237PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Hanley DF, Thompson RE, Rosenblum M et al (2019) Efficacy and safety of minimally invasive surgery with thrombolysis in intracerebral haemorrhage evacuation (MISTIE III): a randomised, controlled, open-label, blinded endpoint phase 3 trial. Lancet. 393(10175):1021–1032PubMedPubMedCentralCrossRef Hanley DF, Thompson RE, Rosenblum M et al (2019) Efficacy and safety of minimally invasive surgery with thrombolysis in intracerebral haemorrhage evacuation (MISTIE III): a randomised, controlled, open-label, blinded endpoint phase 3 trial. Lancet. 393(10175):1021–1032PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Hersh EH, Gologorsky Y, Chartrain AG, Mocco J, Kellner CP (2018) Minimally invasive surgery for intracerebral hemorrhage. Curr Neurol Neurosci Rep 18(6):34PubMedCrossRef Hersh EH, Gologorsky Y, Chartrain AG, Mocco J, Kellner CP (2018) Minimally invasive surgery for intracerebral hemorrhage. Curr Neurol Neurosci Rep 18(6):34PubMedCrossRef
18.
Zurück zum Zitat Hilkens NA, van Asch CJJ, Werring DJ et al (2018) Predicting the presence of macrovascular causes in non-traumatic intracerebral haemorrhage: the DIAGRAM prediction score. J Neurol Neurosurg Psychiatry 89(7):674–679PubMedCrossRef Hilkens NA, van Asch CJJ, Werring DJ et al (2018) Predicting the presence of macrovascular causes in non-traumatic intracerebral haemorrhage: the DIAGRAM prediction score. J Neurol Neurosurg Psychiatry 89(7):674–679PubMedCrossRef
19.
Zurück zum Zitat Hsu CH, Chou SC, Kuo LT et al (2022) Minimally invasive neurosurgery for spontaneous intracerebral hemorrhage-10 years of working progress at National Taiwan University Hospital. Front Neurol 13:817386PubMedPubMedCentralCrossRef Hsu CH, Chou SC, Kuo LT et al (2022) Minimally invasive neurosurgery for spontaneous intracerebral hemorrhage-10 years of working progress at National Taiwan University Hospital. Front Neurol 13:817386PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Keep RF, Hua Y, Xi G (2012) Intracerebral haemorrhage: mechanisms of injury and therapeutic targets. Lancet Neurol 11(8):720–731PubMedCrossRef Keep RF, Hua Y, Xi G (2012) Intracerebral haemorrhage: mechanisms of injury and therapeutic targets. Lancet Neurol 11(8):720–731PubMedCrossRef
21.
Zurück zum Zitat Kellner CP, Chartrain AG, Nistal DA et al (2018) The Stereotactic Intracerebral Hemorrhage Underwater Blood Aspiration (SCUBA) technique for minimally invasive endoscopic intracerebral hemorrhage evacuation. J Neurointerv Surg 10(8):771–776PubMedPubMedCentralCrossRef Kellner CP, Chartrain AG, Nistal DA et al (2018) The Stereotactic Intracerebral Hemorrhage Underwater Blood Aspiration (SCUBA) technique for minimally invasive endoscopic intracerebral hemorrhage evacuation. J Neurointerv Surg 10(8):771–776PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Kellner CP, Song R, Ali M et al (2021) Time to evacuation and functional outcome after minimally invasive endoscopic intracerebral hemorrhage evacuation. Stroke. 52(9):e536–e539PubMedCrossRef Kellner CP, Song R, Ali M et al (2021) Time to evacuation and functional outcome after minimally invasive endoscopic intracerebral hemorrhage evacuation. Stroke. 52(9):e536–e539PubMedCrossRef
23.
Zurück zum Zitat Kellner CP, Song R, Pan J et al (2020) Long-term functional outcome following minimally invasive endoscopic intracerebral hemorrhage evacuation. J Neurointerv Surg 12(5):489–494PubMedPubMedCentralCrossRef Kellner CP, Song R, Pan J et al (2020) Long-term functional outcome following minimally invasive endoscopic intracerebral hemorrhage evacuation. J Neurointerv Surg 12(5):489–494PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Krishnamurthi RV, Ikeda T, Feigin VL (2020) Global, regional and country-specific burden of ischaemic stroke, intracerebral haemorrhage and subarachnoid haemorrhage: a systematic analysis of the Global Burden of Disease Study 2017. Neuroepidemiology. 54(2):171–179PubMedCrossRef Krishnamurthi RV, Ikeda T, Feigin VL (2020) Global, regional and country-specific burden of ischaemic stroke, intracerebral haemorrhage and subarachnoid haemorrhage: a systematic analysis of the Global Burden of Disease Study 2017. Neuroepidemiology. 54(2):171–179PubMedCrossRef
25.
Zurück zum Zitat Kuo LT, Chen CM, Li CH et al (2011) Early endoscope-assisted hematoma evacuation in patients with supratentorial intracerebral hemorrhage: case selection, surgical technique, and long-term results. Neurosurg Focus 30(4):E9PubMedCrossRef Kuo LT, Chen CM, Li CH et al (2011) Early endoscope-assisted hematoma evacuation in patients with supratentorial intracerebral hemorrhage: case selection, surgical technique, and long-term results. Neurosurg Focus 30(4):E9PubMedCrossRef
26.
Zurück zum Zitat Li Y, Wang J, Li Z et al (2018) Computed tomography angiography spot sign as an indicator for ultra-early stereotactic aspiration of intracerebral hemorrhage. World Neurosurg 109:e136–e143PubMedCrossRef Li Y, Wang J, Li Z et al (2018) Computed tomography angiography spot sign as an indicator for ultra-early stereotactic aspiration of intracerebral hemorrhage. World Neurosurg 109:e136–e143PubMedCrossRef
27.
Zurück zum Zitat Li Y, Yang S, Zhou X, Lai R, Tan D (2022) A retrospective cohort study of neuroendoscopic surgery versus traditional craniotomy on surgical success rate, postoperative complications, and prognosis in patients with acute intracerebral hemorrhage. Comput Intell Neurosci 2022:2650795PubMedPubMedCentral Li Y, Yang S, Zhou X, Lai R, Tan D (2022) A retrospective cohort study of neuroendoscopic surgery versus traditional craniotomy on surgical success rate, postoperative complications, and prognosis in patients with acute intracerebral hemorrhage. Comput Intell Neurosci 2022:2650795PubMedPubMedCentral
28.
Zurück zum Zitat Ma L, Hou Y, Zhu R, Chen X (2017) Endoscopic evacuation of basal ganglia hematoma: surgical technique, outcome, and learning curve. World Neurosurg 101:57–68PubMedCrossRef Ma L, Hou Y, Zhu R, Chen X (2017) Endoscopic evacuation of basal ganglia hematoma: surgical technique, outcome, and learning curve. World Neurosurg 101:57–68PubMedCrossRef
29.
Zurück zum Zitat McCulloch P, Altman DG, Campbell WB et al (2009) No surgical innovation without evaluation: the IDEAL recommendations. Lancet 374(9695):1105–1112PubMedCrossRef McCulloch P, Altman DG, Campbell WB et al (2009) No surgical innovation without evaluation: the IDEAL recommendations. Lancet 374(9695):1105–1112PubMedCrossRef
30.
Zurück zum Zitat Mendelow AD, Gregson BA, Fernandes HM et al (2005) Early surgery versus initial conservative treatment in patients with spontaneous supratentorial intracerebral haematomas in the International Surgical Trial in Intracerebral Haemorrhage (STICH): a randomised trial. Lancet. 365(9457):387–397PubMedCrossRef Mendelow AD, Gregson BA, Fernandes HM et al (2005) Early surgery versus initial conservative treatment in patients with spontaneous supratentorial intracerebral haematomas in the International Surgical Trial in Intracerebral Haemorrhage (STICH): a randomised trial. Lancet. 365(9457):387–397PubMedCrossRef
31.
Zurück zum Zitat Mendelow AD, Gregson BA, Rowan EN et al (2013) Early surgery versus initial conservative treatment in patients with spontaneous supratentorial lobar intracerebral haematomas (STICH II): a randomised trial. Lancet. 382(9890):397–408PubMedPubMedCentralCrossRef Mendelow AD, Gregson BA, Rowan EN et al (2013) Early surgery versus initial conservative treatment in patients with spontaneous supratentorial lobar intracerebral haematomas (STICH II): a randomised trial. Lancet. 382(9890):397–408PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Miki K, Yagi K, Nonaka M et al (2018a) Spot sign as a predictor of rebleeding after endoscopic surgery for intracerebral hemorrhage. J Neurosurg:1–6 Miki K, Yagi K, Nonaka M et al (2018a) Spot sign as a predictor of rebleeding after endoscopic surgery for intracerebral hemorrhage. J Neurosurg:1–6
33.
Zurück zum Zitat Miki K, Yagi K, Nonaka M et al (2018b) Intraoperative active bleeding in endoscopic surgery for spontaneous intracerebral hemorrhage is predicted by the spot sign. World Neurosurg 116:e513–e518PubMedCrossRef Miki K, Yagi K, Nonaka M et al (2018b) Intraoperative active bleeding in endoscopic surgery for spontaneous intracerebral hemorrhage is predicted by the spot sign. World Neurosurg 116:e513–e518PubMedCrossRef
34.
Zurück zum Zitat Morgenstern LB, Demchuk AM, Kim DH, Frankowski RF, Grotta JC (2001) Rebleeding leads to poor outcome in ultra-early craniotomy for intracerebral hemorrhage. Neurology. 56(10):1294–1299PubMedCrossRef Morgenstern LB, Demchuk AM, Kim DH, Frankowski RF, Grotta JC (2001) Rebleeding leads to poor outcome in ultra-early craniotomy for intracerebral hemorrhage. Neurology. 56(10):1294–1299PubMedCrossRef
35.
Zurück zum Zitat Mould WA, Carhuapoma JR, Muschelli J et al (2013) Minimally invasive surgery plus recombinant tissue-type plasminogen activator for intracerebral hemorrhage evacuation decreases perihematomal edema. Stroke. 44(3):627–634PubMedPubMedCentralCrossRef Mould WA, Carhuapoma JR, Muschelli J et al (2013) Minimally invasive surgery plus recombinant tissue-type plasminogen activator for intracerebral hemorrhage evacuation decreases perihematomal edema. Stroke. 44(3):627–634PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Pan J, Chartrain AG, Scaggiante J et al (2020) A compendium of modern minimally invasive intracerebral hemorrhage evacuation techniques. Oper Neurosurg 18(6):710–720CrossRef Pan J, Chartrain AG, Scaggiante J et al (2020) A compendium of modern minimally invasive intracerebral hemorrhage evacuation techniques. Oper Neurosurg 18(6):710–720CrossRef
37.
Zurück zum Zitat Pantazis G, Tsitsopoulos P, Mihas C, Katsiva V, Stavrianos V, Zymaris S (2006) Early surgical treatment vs conservative management for spontaneous supratentorial intracerebral hematomas: a prospective randomized study. Surg Neurol 66(5):492–501 discussion 501-492PubMedCrossRef Pantazis G, Tsitsopoulos P, Mihas C, Katsiva V, Stavrianos V, Zymaris S (2006) Early surgical treatment vs conservative management for spontaneous supratentorial intracerebral hematomas: a prospective randomized study. Surg Neurol 66(5):492–501 discussion 501-492PubMedCrossRef
38.
Zurück zum Zitat Poon MT, Fonville AF, Al-Shahi SR (2014) Long-term prognosis after intracerebral haemorrhage: systematic review and meta-analysis. J Neurol Neurosurg Psychiatry 85(6):660–667PubMedCrossRef Poon MT, Fonville AF, Al-Shahi SR (2014) Long-term prognosis after intracerebral haemorrhage: systematic review and meta-analysis. J Neurol Neurosurg Psychiatry 85(6):660–667PubMedCrossRef
39.
Zurück zum Zitat Rabinstein AA, Anderson CD (2015) Time is brain also counts for ICH. Neurology. 84(10):970–971PubMedCrossRef Rabinstein AA, Anderson CD (2015) Time is brain also counts for ICH. Neurology. 84(10):970–971PubMedCrossRef
40.
41.
Zurück zum Zitat Rothrock RJ, Chartrain AG, Scaggiante J et al (2021) Advanced techniques for endoscopic intracerebral hemorrhage evacuation: a technical report with case examples. Oper Neurosurg 20(1):119–129CrossRef Rothrock RJ, Chartrain AG, Scaggiante J et al (2021) Advanced techniques for endoscopic intracerebral hemorrhage evacuation: a technical report with case examples. Oper Neurosurg 20(1):119–129CrossRef
42.
Zurück zum Zitat Ruiz-Sandoval JL, Chiquete E, Romero-Vargas S, Padilla-Martinez JJ, Gonzalez-Cornejo S (2007) Grading scale for prediction of outcome in primary intracerebral hemorrhages. Stroke. 38(5):1641–1644PubMedCrossRef Ruiz-Sandoval JL, Chiquete E, Romero-Vargas S, Padilla-Martinez JJ, Gonzalez-Cornejo S (2007) Grading scale for prediction of outcome in primary intracerebral hemorrhages. Stroke. 38(5):1641–1644PubMedCrossRef
43.
Zurück zum Zitat Scaggiante J, Zhang X, Mocco J, Kellner CP (2018) Minimally invasive surgery for intracerebral hemorrhage. Stroke. 49(11):2612–2620PubMedCrossRef Scaggiante J, Zhang X, Mocco J, Kellner CP (2018) Minimally invasive surgery for intracerebral hemorrhage. Stroke. 49(11):2612–2620PubMedCrossRef
44.
Zurück zum Zitat Sondag L, Schreuder F, Boogaarts HD et al (2020) Neurosurgical intervention for supratentorial intracerebral hemorrhage. Ann Neurol 88(2):239–250PubMedPubMedCentralCrossRef Sondag L, Schreuder F, Boogaarts HD et al (2020) Neurosurgical intervention for supratentorial intracerebral hemorrhage. Ann Neurol 88(2):239–250PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Song R, Ali M, Smith C et al (2022) Initial experience with the NICO myriad device for minimally invasive endoscopic evacuation of intracerebral hemorrhage. Oper Neurosurg 23(3):194–199CrossRef Song R, Ali M, Smith C et al (2022) Initial experience with the NICO myriad device for minimally invasive endoscopic evacuation of intracerebral hemorrhage. Oper Neurosurg 23(3):194–199CrossRef
46.
Zurück zum Zitat Spiotta AM, Fiorella D, Vargas J et al (2015) Initial multicenter technical experience with the Apollo device for minimally invasive intracerebral hematoma evacuation. Neurosurgery. 11(Suppl 2):243–251 discussion 251PubMed Spiotta AM, Fiorella D, Vargas J et al (2015) Initial multicenter technical experience with the Apollo device for minimally invasive intracerebral hematoma evacuation. Neurosurgery. 11(Suppl 2):243–251 discussion 251PubMed
47.
Zurück zum Zitat Sun GC, Chen XL, Hou YZ et al (2017) Image-guided endoscopic surgery for spontaneous supratentorial intracerebral hematoma. J Neurosurg 127(3):537–542PubMedCrossRef Sun GC, Chen XL, Hou YZ et al (2017) Image-guided endoscopic surgery for spontaneous supratentorial intracerebral hematoma. J Neurosurg 127(3):537–542PubMedCrossRef
48.
Zurück zum Zitat van Asch CJ, Luitse MJ, Rinkel GJ, van der Tweel I, Algra A, Klijn CJ (2010) Incidence, case fatality, and functional outcome of intracerebral haemorrhage over time, according to age, sex, and ethnic origin: a systematic review and meta-analysis. Lancet Neurol 9(2):167–176PubMedCrossRef van Asch CJ, Luitse MJ, Rinkel GJ, van der Tweel I, Algra A, Klijn CJ (2010) Incidence, case fatality, and functional outcome of intracerebral haemorrhage over time, according to age, sex, and ethnic origin: a systematic review and meta-analysis. Lancet Neurol 9(2):167–176PubMedCrossRef
49.
Zurück zum Zitat van Asch CJ, Velthuis BK, Rinkel GJ et al (2015) Diagnostic yield and accuracy of CT angiography, MR angiography, and digital subtraction angiography for detection of macrovascular causes of intracerebral haemorrhage: prospective, multicentre cohort study. BMJ. 351:h5762PubMedPubMedCentralCrossRef van Asch CJ, Velthuis BK, Rinkel GJ et al (2015) Diagnostic yield and accuracy of CT angiography, MR angiography, and digital subtraction angiography for detection of macrovascular causes of intracerebral haemorrhage: prospective, multicentre cohort study. BMJ. 351:h5762PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Vespa P, Hanley D, Betz J et al (2016) ICES (intraoperative stereotactic computed tomography-guided endoscopic surgery) for brain hemorrhage: a multicenter randomized controlled trial. Stroke. 47(11):2749–2755PubMedPubMedCentralCrossRef Vespa P, Hanley D, Betz J et al (2016) ICES (intraoperative stereotactic computed tomography-guided endoscopic surgery) for brain hemorrhage: a multicenter randomized controlled trial. Stroke. 47(11):2749–2755PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Volbers B, Willfarth W, Kuramatsu JB et al (2016) Impact of perihemorrhagic edema on short-term outcome after intracerebral hemorrhage. Neurocrit Care 24(3):404–412PubMedCrossRef Volbers B, Willfarth W, Kuramatsu JB et al (2016) Impact of perihemorrhagic edema on short-term outcome after intracerebral hemorrhage. Neurocrit Care 24(3):404–412PubMedCrossRef
52.
Zurück zum Zitat Xia Z, Wu X, Li J et al (2018) Minimally invasive surgery is superior to conventional craniotomy in patients with spontaneous supratentorial intracerebral hemorrhage: a systematic review and meta-analysis. World Neurosurg 115:266–273PubMedCrossRef Xia Z, Wu X, Li J et al (2018) Minimally invasive surgery is superior to conventional craniotomy in patients with spontaneous supratentorial intracerebral hemorrhage: a systematic review and meta-analysis. World Neurosurg 115:266–273PubMedCrossRef
53.
Zurück zum Zitat Ye Y, Wang Q, Ou W, He J, Zhao Z (2020) Endoscopic surgery without decompressive craniectomy in large putaminal intracerebral hemorrhage: assessment of efficacy and safety. Neurocrit Care 32(2):392–399PubMedCrossRef Ye Y, Wang Q, Ou W, He J, Zhao Z (2020) Endoscopic surgery without decompressive craniectomy in large putaminal intracerebral hemorrhage: assessment of efficacy and safety. Neurocrit Care 32(2):392–399PubMedCrossRef
54.
Zurück zum Zitat Yushkevich PA, Piven J, Hazlett HC et al (2006) User-guided 3D active contour segmentation of anatomical structures: significantly improved efficiency and reliability. Neuroimage. 31(3):1116–1128PubMedCrossRef Yushkevich PA, Piven J, Hazlett HC et al (2006) User-guided 3D active contour segmentation of anatomical structures: significantly improved efficiency and reliability. Neuroimage. 31(3):1116–1128PubMedCrossRef
55.
Zurück zum Zitat Zhou X, Xie L, Altinel Y, Qiao N (2020) Assessment of evidence regarding minimally invasive surgery vs. conservative treatment on intracerebral hemorrhage: a trial sequential analysis of randomized controlled trials. Front Neurol 11:426PubMedPubMedCentralCrossRef Zhou X, Xie L, Altinel Y, Qiao N (2020) Assessment of evidence regarding minimally invasive surgery vs. conservative treatment on intracerebral hemorrhage: a trial sequential analysis of randomized controlled trials. Front Neurol 11:426PubMedPubMedCentralCrossRef
Metadaten
Titel
Safety and technical efficacy of early minimally invasive endoscopy-guided surgery for intracerebral haemorrhage: the Dutch Intracerebral haemorrhage Surgery Trial pilot study
verfasst von
Lotte Sondag
Floris H.B.M. Schreuder
Sjoert A.H. Pegge
Jonathan M. Coutinho
Diederik W.J. Dippel
Paula M. Janssen
W. Peter Vandertop
Hieronymus D. Boogaarts
Ruben Dammers
Catharina J.M. Klijn
On behalf of the Dutch ICH Surgery Trial Study Group, part of the CONTRAST consortium
Publikationsdatum
27.04.2023
Verlag
Springer Vienna
Erschienen in
Acta Neurochirurgica / Ausgabe 6/2023
Print ISSN: 0001-6268
Elektronische ISSN: 0942-0940
DOI
https://doi.org/10.1007/s00701-023-05599-2

Weitere Artikel der Ausgabe 6/2023

Acta Neurochirurgica 6/2023 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Sozialer Aufstieg verringert Demenzgefahr

24.05.2024 Demenz Nachrichten

Ein hohes soziales Niveau ist mit die beste Versicherung gegen eine Demenz. Noch geringer ist das Demenzrisiko für Menschen, die sozial aufsteigen: Sie gewinnen fast zwei demenzfreie Lebensjahre. Umgekehrt steigt die Demenzgefahr beim sozialen Abstieg.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Was nützt die Kraniektomie bei schwerer tiefer Hirnblutung?

17.05.2024 Hirnblutung Nachrichten

Eine Studie zum Nutzen der druckentlastenden Kraniektomie nach schwerer tiefer supratentorieller Hirnblutung deutet einen Nutzen der Operation an. Für überlebende Patienten ist das dennoch nur eine bedingt gute Nachricht.

Thrombektomie auch bei großen Infarkten von Vorteil

16.05.2024 Ischämischer Schlaganfall Nachrichten

Auch ein sehr ausgedehnter ischämischer Schlaganfall scheint an sich kein Grund zu sein, von einer mechanischen Thrombektomie abzusehen. Dafür spricht die LASTE-Studie, an der Patienten und Patientinnen mit einem ASPECTS von maximal 5 beteiligt waren.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.