|
THE MAGIC-MT TRIAL
|
♦
Study objective
Primary Objective:
Incidence of recurrence or progression of hematoma within 90 days after surgery
Hematoma recurrence refers that the maximum thickness of hematoma of patients in the operation group exceeds 10 mm, combined with neurological symptoms, or that the patient needs to undergo re-operation;
Symptomatic chronic hematoma progression refers that the maximum thickness of hematoma of patients in the non-operation group increases by more than 3 mm compared with baseline, or the patient needs to receive operation.
Secondary Objective:
(1). Effectiveness [ Time Frame: 1 year post-procedure ]
Incidence of SDH recurrence/ progression at 1 year post-procedure
(2). Effectiveness [ Time Frame: day 0 ]
Rate of successful embolization of the target vessels (MMA trunk and branches) with ONYX base on DSA imaging
(3). Effectiveness [ Time Frame: 90 days post-procedure ]
Change in hematoma thickness based on CT/MRI imaging at 90 days post-procedure
(4). Effectiveness [ Time Frame: 90 days post-procedure ]
Changes in hematoma volume at 90 days post-procedure
(5). Effectiveness [ Time Frame: 90 days post-procedure ]
Change in Midline shift based on CT/MRI imaging at 90 days post-procedure
(6). Effectiveness [ Time Frame: 90 days and 1 year post-procedure ]
Change in the Modified Rankin Scale score (mRS) Grade 0 (no symptoms) to 6 (death) at 90 days and 1 year post-procedure
(7). Effectiveness [ Time Frame: 90 days and 1 year post-procedure ]
Percentage of patients with favorable functional outcome defined as Modified Rankin Scale of 0 to 3 at 90 days and 1year post-procedure
(8). Effectiveness [ Time Frame: 90 days and 1 year post-procedure ]
Percentage of patients with good functional outcome defined as Modified Rankin Scale of 0 to 2 at 90 days and 1 year post-procedure
(9). Effectiveness [ Time Frame: 90 days and 1 year post-procedure ]
Quality of life assessed by (EuroQol) EQ-5D scale Grade 0 (worst health) to 100 (best health) at 90 days and 1 year post-procedure
(10). Safety endpoint [ Time Frame: 90 days ]
Total patients with SAEs within 90 days post-procedure
(11). Safety endpoint [ Time Frame: 90 days ]
Incidence of neurological death within 90 days post-procedure
(12). Safety endpoint [ Time Frame: 30 days ]
Incidence of procedural serious complications within 30 days post-procedure:
o symptomatic procedure-related intracranial hemorrhage
o any procedure-related intracranial hemorrhage
o any procedure-related neurological deficit
o CNS infection caused by procedure
o procedure-related artery dissection, vessel wall damage and vessel perforation
o procedure-related ischemic event
o retroperitoneal hematoma (femoral access)/wrist hematoma (radial access)
o neuropathy at the puncture site
o contrast agent allergy or encephalopathy
♦
Study design
Study Type : | Interventional (Clinical Trial) |
Estimated Enrollment : | 722 participants |
Allocation : | Randomized |
Intervention Model : | Parallel Assignment |
Masking : | None (Open Label) |
Primary Purpose : | Treatment |
Official Title : | Managing Non-acute Subdural Hematoma Using Liquid Materials:a Chinese Randomized Trial of MMA Treatment |
♦
Study population
Patients with symptomatic non-acute SDH with mass effect. Age must be 18 or higher and mRS score must be ≤ 2 prior to the symptom.
♦
Inclusion criteria
1. Symptomatic non-acute SDH with mass effect
Symptomatic SDH is defined as an SDH that fulfills one or more of the following criteria AND shows neurological symptoms (headache, short-term cognitive decline, speech difficulty or aphasia, gait impairment, focal weakness, sensory deficits, seizures) that are attributable to the hematoma
2. Age 18 or higher
3. Independent functional status in activities of daily living with mRS score ≤ 2 prior to the symptom.
4. Signed informed consent or appropriate signed deferral of consent where approved.
♦
Exclusion criteria
1. Qualifying imaging reveals clear hypodense demarcation of the majority of the territory of symptomatic intracranial infarction.
2. Subject requires (in the opinion of the treating surgeon) a full or mini craniotomy.
3. Subject with urgent or emergent subdural hematoma evacuation needed.
4. Bilateral SDH with unknown origin of symptoms.
5. Any evidence of anatomical variants that might render safe MMA embolization impossible (e.g. prominent MMA-ophtalmic artery anastomoses)
6. Coagulation and platelet disorders, or antiplatelet medication that is not easily correctable with INR >1.5 and/or platelet count < 80.000.
7. Contraindications for neurangiography, e.g. iodinated contrast allergy, renal insufficiency with GFR < 30 ml/min.
8. Contraindications for neurangiography, e.g. iodinated contrast allergy, renal insufficiency with GFR < 30 ml/min.
9. Pregnancy; if a woman is of child-bearing potential a urine or serum beta HCG test is positive.
10. Patient has a severe or fatal comorbid illness that will prevent improvement or follow-up or that will render the procedure unlikely to benefit the patient.
11. Life expectancy <1 year.
12. High risk of removal from antiplatelet and/or anticoagulant medical therapy for recent procedure or other reasons.
13. Patient cannot complete follow-up for any reason. (eg. visiting from another city, illness, incarceration in prison etc.)
14. Participation in another clinical trial investigating a drug, medical device, or a medical procedure that might confound treatment and outcomes related to the current trial.
15. Previous surgery or endovascular treatment for the SDH under investigation
16. Unable to undergo MMA embolization prior to surgical treatment
♦
Study procedures
After enrollment, the patient is followed up 4 times:
o Total hospital stay, destination for rehabilitation after discharge and disease treatment and nursing expenses are recorded at discharge
o Vital signs of the 30-day postoperative visit
o Re-examine cranial CT/ MRI 90 days after surgery, and record total hospital stay, re-hospitalization times,destination for rehabilitation after discharge and disease treatment and nursing expenses
o Re-examine cranial CT/ MRI 360 days after surgery, and record total hospital stay, re-hospitalization times,destination for rehabilitation after discharge and disease treatment and nursing expenses
♦
Literature
(1). A. Srivatsan, A. Mohanty, F.A. Nascimento, M.U. Hafeez, V.M. Srinivasan, A. Thomas, S.R. Chen, J.N. Johnson, P. Kan, Middle Meningeal Artery Embolization for Chronic Subdural Hematoma: Meta-Analysis and Systematic Review, World Neurosurg, 122 (2019) 613-619.
(2). T.W. Link, S. Boddu, S.M. Paine, H. Kamel, J. Knopman, Middle Meningeal Artery Embolization for Chronic Subdural Hematoma: A Series of 60 Cases, Neurosurgery, 85 (2019) 801-807.
(3). T.W. Link, B.I. Rapoport, S.M. Paine, H. Kamel, J. Knopman, Middle meningeal artery embolization for chronic subdural hematoma: Endovascular technique and radiographic findings, Interv Neuroradiol, 24 (2018) 455-462.
(4). T.W. Link, J.T. Schwarz, S.M. Paine, H. Kamel, J. Knopman, Middle Meningeal Artery Embolization for Recurrent Chronic Subdural Hematoma: A Case Series, World Neurosurg, 118 (2018) e570-e574.
(5). R. Tang, J. Shi, X. Li, Y. Zou, L. Wang, Y. Chen, R. Yan, B. Gao, H. Feng, Effects of Atorvastatin on Surgical Treatments of Chronic Subdural Hematoma, World Neurosurg, 117 (2018) e425-e429.
(6). W. Lu, H. Wang, T. Wu, X. Sheng, Z. Ding, G. Xu, Burr-Hole Craniostomy with T-Tube Drainage as Surgical Treatment for Chronic Subdural Hematoma, World Neurosurg, 115 (2018) e756-e760.
(7). R. Jiang, S. Zhao, R. Wang, H. Feng, J. Zhang, X. Li, Y. Mao, X. Yuan, Z. Fei, Y. Zhao, X. Yu, W.S. Poon, X. Zhu, N. Liu, D. Kang, T. Sun, B. Jiao, X. Liu, R. Yu, J. Zhang, G. Gao, J. Hao, N. Su, G. Yin, X. Zhu, Y. Lu, J. Wei, J. Hu, R. Hu, J. Li, D. Wang, H. Wei, Y. Tian, P. Lei, J.F. Dong, J. Zhang, Safety and Efficacy of Atorvastatin for Chronic Subdural Hematoma in Chinese Patients: A Randomized ClinicalTrial, JAMA Neurol, 75 (2018) 1338-1346.
(8). J. Van Der Veken, J. Duerinck, R. Buyl, K. Van Rompaey, P. Herregodts, J. D'Haens, Mini-craniotomy as the primary surgical intervention for the treatment of chronic subdural hematoma--a retrospective analysis, Acta Neurochir (Wien), 156 (2014) 981-987.
(9). J. Yu, Y. Guo, B. Xu, K. Xu, Clinical importance of the middle meningeal artery: A review of the literature, Int J Med Sci, 13 (2016) 790-799.
(10).K. Takahashi, K. Muraoka, T. Sugiura, Y. Maeda, S. Mandai, Y. Gohda, M. Kawauchi, Y. Matsumoto, [Middle meningeal artery embolization for refractory chronic subdural hematoma: 3 case reports], No Shinkei Geka, 30 (2002) 535-539.
(11).T. Tanaka, S. Fujimoto, K. Saitoh, S. Satoh, K. Nagamatsu, H. Midorikawa, [Superselective angiographic findings of ipsilateral middle meningeal artery of chronic subdural hematoma in adults], No Shinkei Geka, 26 (1998) 339-347.
(12).F. Altinel, C. Altin, E. Gezmis, N. Altinors, Cortical membranectomy in chronic subdural hematoma: Report of two cases, Asian J Neurosurg, 10 (2015) 236-239.
(13).M. Suzuki, S. Endo, K. Inada, A. Kudo, A. Kitakami, K. Kuroda, A. Ogawa, Inflammatory cytokines locally elevated in chronic subdural haematoma, Acta Neurochir (Wien), 140 (1998) 51-55.
(14).S. Mandai, M. Sakurai, Y. Matsumoto, Middle meningeal artery embolization for refractory chronic subdural hematoma. Case report, J Neurosurg, 93 (2000) 686-688.
(15).D.C. Holl, V. Volovici, C.M.F. Dirven, W.C. Peul, F. van Kooten, K. Jellema, N.A. van der Gaag, I.P. Miah, K.H. Kho, H.M. den Hertog, H.F. Lingsma, R. Dammers, G. Dutch Chronic Subdural Hematoma Research, Pathophysiology and Nonsurgical Treatment of Chronic Subdural Hematoma: From Past to Present to Future, World Neurosurg, 116 (2018) 402-411 e402.
(16).M. Haldrup, B. Ketharanathan, B. Debrabant, O.S. Schwartz, R. Mikkelsen, K. Fugleholm, F.R. Poulsen, T.S.R. Jensen, L.V. Thaarup, B. Bergholt, Embolization of the middle meningeal artery in patients with chronic subdural hematoma-a systematic review and meta-analysis, Acta Neurochir (Wien), 162 (2020) 777-784.
(17).S.P. Ban, G. Hwang, H.S. Byoun, T. Kim, S.U. Lee, J.S. Bang, J.H. Han, C.Y. Kim, O.K. Kwon, C.W. Oh, Middle Meningeal Artery Embolization for Chronic Subdural Hematoma, Radiology, 286 (2018) 992-999.
(18).A.J. Molyneux, S. Cekirge, I. Saatci, G. Gal, Cerebral Aneurysm Multicenter European Onyx (CAMEO) trial: results of a prospective observational study in 20 European centers, AJNR Am J Neuroradiol, 25 (2004) 39-51.
(19).G.B. Rajah, M. Waqas, R.H. Dossani, K. Vakharia, A.D. Gong, K. Rho, S.B. Housley, H.H. Rai, F. Chin, M.K. Tso, K.V. Snyder, E.I. Levy, A.H. Siddiqui, J.M. Davies, Transradial middle meningeal artery embolization for chronic subdural hematoma using Onyx: case series, J Neurointerv Surg, DOI 10.1136/neurintsurg-2020- 016185(2020).
♦
Statistical analysis plan
Title : |
Managing Non-acute Subdural Hematoma Using Liquid Materials:
a Chinese Randomized Trial of MMA Treatment |
Protocol No. : |
MAGIC-MT |
Version No. : |
V1.0 |
Effective Date : |
2023-12-02 |
Description:
• The purpose of this Statistical Analysis Plan (SAP) is to describe the planned analyses and
presentation of results in the clinical study report corresponding to the study protocol
MAGIC-MT .
• The purpose of this SAP is to describe the efficacy analysis and safety analysis required
by the study protocol for middle meningeal artery (MMA) embolization using liquid
embolic material for the treatment of non-acute subdural hematoma.
• This SAP will be provided to study team members to communicate the full delivery of the
statistical analysis.
Author name, affiliation and position:
Liang Zhou
dMed Biopharmaceutical Co., Ltd.
Senior Biostatistician
APPROVED BY:
Name, affiliation and position
|
Signature
|
Date
|
Ying Mao
Huashan Hospital, Fudan University
Professor
|
|
|
Name, affiliation and position
|
Signature
|
Date
|
Jianmin Liu
First Affiliated Hospital of the Second
Military Medical University (Changhai
Hospital of Shanghai)
Professor
|
|
|
Version History
This SAP is based on the MAGIC-MT study protocol version 3.0 version dated 2022-09-21 .
SAP Version
|
Effective Date
|
Author
|
Revision Description
|
1.0
|
2023-12-02
|
Zhou Liang
|
Original Version
|
CONTENTS
Version History
1. Summary of Protocol
1.1. Statistical Analysis Plan and Protocol Discrepancies
1.2. Study Objectives and Endpoints
1.3. Study Design
2. Statistical Hypothesis
3. Sample Size Considerations
4. Planned Analyses
4.1. Interim Analyses
4.2. Final Analysis
5. Analysis Population
5.1. Protocol Deviations
6. Considerations for Data Analyses and Data Handling
Conventions
7. Study Population Analyses
7.1. Overview of Planned Study Population Analyses
7.2. Description Of Study Population Analysis
7.2.1. Demographic and Baseline Characteristics
8. Primary Efficacy Endpoints Analyses
8.1. Overview of Planned Primary Efficacy Endpoints Analyses
8.2. Specifications of Primary Efficacy Endpoints Analyses
8.2.1. Definitions and Derivations
8.2.2. Analysis Methods
8.2.3. Model Specifications
8.3. Subgroup Analyses
9. Secondary Statistical Analyses
9.1. Secondary Efficacy Endpoint Analyses
9.1.1. Overview of Planned Analyses for Secondary Efficacy Endpoints
9.1.2. Specifications of Secondary Efficacy Endpoint Analyses
9.2 Safety analyses
9.2.1 Overview of Planned Safety Analyses
9.2.2 Specifications Of Safety Analyses
9.2.2.1 Adverse Events
10. References
12. APPENDICES
1 2.1 Appendix 1: Treatment Period and Study Period
12.2 Appendix 2: Data Display Standards and Handling Conventions
12.3 Appendix 3: Handling of Early Withdrawals and Missing Data
12.4 Appendix 4: Abbreviations Table
1. Summary of Protocol
1.1. Statistical Analysis Plan and Protocol Discrepancies
All changes to the content of the analyses originally specified in Protocol 3.0 (2022-09-21) are
summarized in Table 1.
Table 1 Changes to the analysis plan defined in the protoco
Protocol
|
Statistical Analysis Plan
|
|
Statistical description
in the protocol
|
Statistical description in
statistical analysis plan
|
Reason for change
|
• Section 7.3.1 For the
primary endpoint,
CMH chi-square
analysis will be used
to calculate the
difference in the
event rate between
the two groups and
its 95% confidence
interval, and KaplanMeier method will be
further used to
estimate the event
rate of the primary
endpoint and its 95%
confidence interval.
|
• Section 8.2.2 Primary
analysis:90-day recurrence
or progression rates will be
summarized for each group
using a two-sided 95% CI
(Clopper-Pearson method).
P-values for differences in
recurrence or progression
rates between the 2 groups
will be compared between
groups using the CMH chisquare test. The stratification
factor is whether or not
trepanation and drainage is
performed. In addition, a
two-sided 95% CI for the
difference in recurrence or
progression rates between
the two groups will be
calculated using the
Miettinen-Nurminen
method.
• Sensitivity analysis:
Generalized linear mixedeffects model will be used to
calculate ORs and their 95%
CIs for the comparison of
recurrence or progression
rates between the two
groups for sensitivity
analysis with group as a
covariate, site as a random
effect. Multivariable
analysis will also be
provided to include group,
sex, age, maximum
thickness of hematoma on
the enrolled side, baseline
mRS score.
|
• According to internal
discussion as well as
reference to the design of
similar studies, time-toevent analyses were not
included for the primary
endpoint, and logistic
regression analysis methods
were added.
|
• Section 7.3.2 The
analyses of secondary
endpoints will be the
same as those for the
primary endpoint,
and comparisons
between groups will
be made using the
Chi-square test
(qualitative measure)
or t-test (quantitative
measure). For event
endpoints, the Kaplan
Meier method will be
used.
|
• For two continuous
secondary endpoints,
Change of maximum
hematoma thickness and
hematoma volume between
baseline and 90 days, a
linear mixed effects model
approach will be used to
estimate the mean estimate
(95% CI) of change from
baseline for both treatment
groups and mean estimate
(95% CI) of difference
between groups and p-value
of difference between
groups; Treatment group
variables, sex, age, surgical
treatment strata
(trepanation/drainage vs. no
trepanation/drainage), any
usage of antiplatelet or
anticoagulant therapy within
one month before
randomization(yes vs no),
hematoma volume of
baseline CT will be included
in the model. The random
effects will be site. For other
continuous secondary
endpoints, group
comparisons will be
performed using t-tests or
Mann-Whitney U tests and
95% confidence intervals for
the difference in means
between groups will be
calculated using normal
approximation.
|
• According to internal
discussion as well as
reference to the design of
similar studies, secondary
endpoints do not include
time-to-event analyses and
linear mixed effects model
analysis methods are added.
|
• None
|
• Section 8.2.1 The results of
central imaging assessment
analysis of CT/MRI
examinations required for
the determination of the
subject 's primary endpoint
in this study will be
presented in tables. Central
image evaluation will be
performed by the
Independent Central Image
Review Committee.
Neurological symptoms and
follow-up mRS scores were
derived from OAC review
data.
• Section 9.3.2.1 CEC
assessments for adverse
events of special interest.
|
• Added central imaging
assessments (CT/MRI
examinations), CEC
assessments (adverse events
of special interest), and
clarified data sources.
|
1.2. Study Objectives and Endpoints
Study objectives
|
Study Endpoints
|
Primary objective
|
Primary Study Endpoints
|
• To validate that in patients
with symptomatic non-acute
subdural hematoma (SDH),
MMA embolization with
liquid embolic materials can
reduce a) the incidence of
symptomatic hematoma
recurrence for patients
receiving trepanation and
drainage and b) the incidence
of symptomatic hematoma
progression for patients
undergoing conservative
treatment.
|
• Incidence of symptomatic recurrence (in patients who
undergo trepanation/drainage) or symptomatic
progression of hematoma (in patients who do not
undergo trepanation/drainage) within 90 days after
randomization
• Symptomatic hematoma recurrence is defined as
maximum hematoma thickness >10 mm in patients in
the trepanation group, combined with neurological
symptoms, or that the patient needs to undergo reoperation
• Symptomatic hematoma progression is defined as an
increase of >3mm maximum thickness of SDH of
patients in the non-trepanation group compared with
baseline, or the need for surgery based on the
assessment of the treating medical team
|
Secondary objectives
|
Secondary Study End Point
|
• To validate the efficacy of
MMA embolization with
liquid embolic materials
|
• Incidence of SDH symptomatic recurrence (in
patients who undergo trepanation/drainage) and
symptomatic progression (in patients who do not
undergo trepanation/drainage) 360 days after
randomization
• Success rate of target vessel embolization in DSA
imaging
|
• To validate the safety of
MMA embolization with
liquid embolic materials
|
• Section 8.2.1 The results of
central imaging assessment
analysis of CT/MRI
examinations required for
the determination of the
subject 's primary endpoint
in this study will be
presented in tables. Central
image evaluation will be
performed by the
Independent Central Image
Review Committee.
Neurological symptoms and
follow-up mRS scores were
derived from OAC review
data.
• Section 9.3.2.1 CEC
assessments for adverse
events of special interest.
|
• To validate the reduction of
residual hematoma at 90 days
after randomization in patients
undergoing MMA
embolization
|
• Changes of hematoma thickness on CT/MRI imaging
at 90 days after randomization
• Changes of hematoma volume at 90 days after
randomization
• Changes of midline shift in CT/MRI imaging at 90
days after randomization
|
• To validate the improvement
in clinical outcome (mRS
score) at 90 days and 360 days
after randomization in patients
undergoing MMA
embolization
|
• Changes of Modified Rankin Scale (mRS) score at 90
days and 360 days after randomization
• Proportion of patients with mRS score of 0 to 2 at 90
days and 360 days after randomization
• Proportion of patients with mRS score of 0 to 3 at 90
days and 360 days after randomization
|
• To validate the improvement
in health status (EQ-5D-5L
scale) at 90 days and 360 days
after randomization in patients
undergoing MMA
embolization
|
• Quality of life evaluated with EQ-5D-5L Scale at 90
days and 360 days after randomization
|
• To validate the reduction of
readmission rate within 90
days after randomization in
patients undergoing MMA
embolization
|
• Total days of hospital stay, re-hospitalization times,
destination for rehabilitation after discharge (going
home vs rehabilitation hospital) and total
hospitalization expenses
|
1.3. Study Design
Overview of Study Design and Key Characteristics
|
|
Design
Features
|
• This is a multi-center, randomized, open-label, blinded evaluation
endpoint, and parallel controlled clinical study. The study contents
include: 1) Screening and baseline period: Confirm whether the patients
are eligible for the study and make a record and decide whether
trepanation and drainage should be performed by considering the
condition of the disease and the imaging results, and then carry out
randomization and therapy. Eligible subjects will be randomly assigned
to the traditional treatment group or MMA embolization group at the
ratio of 1:1. 2) Follow-up period: Assess patients for efficacy endpoints
and safety endpoints at 30 days, 90 days and 360 days after
randomization.
|
Study
treatment
|
• MMA embolization: The surgery is performed under local anesthesia +
analgesia, or general anesthesia, and the type of anesthesia is decided by
the neurointerventionalists. Different types of catheters are chosen
according to patients' anatomy. Arterial access is usually attained via
femoral artery groin puncture, however radial or brachial artery access is
also allowed. Angiography will usually show MMA dilatation and
abnormal vascular staining on the affected side, and then embolic
materials are slowly injected into the blood vessels, while avoiding reflux
carefully. Investigators who use liquid embolic materials shall be
qualified in corresponding training before they can perform the surgery.
• Drug therapy: Drug treatment can be divided into the symptomatic
treatment and treatment promoting hematoma absorption. Indications of
drug treatment promoting hematoma absorption: 1) Confirmed diagnosis
of non-acute SDH; 2) Patients complicated with multiple organ failure
and coagulation dysfunction, etc. who are not suitable for surgery or
refuse the surgery; and 3) To prevent postoperative recurrence after
surgical treatment etc. The contraindications are: allergic to the drug used
or having contraindications for the use of the drug.
Atorvastatin calcium and dexamethasone are recommended. Atorvastatin
calcium with small dose and long course of treatment can be used in
combination with dexamethasone. The specific treatment is subject to the
local clinical diagnosis and treatment specifications or corresponding
clinical guidelines, and is not specified in this study.
The application of hemostatic drugs will greatly increase the incidence
risk of embolic diseases, so hemostatic drugs are not commonly used.
• Surgical Treatment: Surgical treatment in this study refers to trepanation
and drainage, and local anesthesia + analgesia or general anesthesia can
be used for trepanation and drainage.
Indications of trepanation and drainage: (1) There are clinical symptoms
and signs of high intracranial pressure, with or without changes in
consciousness and signs of cerebral hemisphere compression; (2) CT or
MRI scan shows unilateral or bilateral subdural hematoma with thickness
of > 10 mm, and midline shift of > 10 mm due to unilateral hematoma;
(3) For those who have been treated with drugs for promoting hematoma
absorption for 2 weeks or longer, and whose clinical manifestations and
imaging examination have not improved significantly, or whose
hematoma continues to increase or medication is intolerable, surgical
treatment is recommended.
If the subjects in the embolization group need surgery, the embolization
will be performed before trepanation and drainage.
|
Assignment
|
• Eligible subjects will be randomized 1:1 to either conventional therapy
(no MMA embolization) or MMA embolization.
|
Interim
Analyses
|
• The study includes two formal interim analyses, and efficacy analyses
will be performed when approximately one-third and two-thirds of
patients completed the 90-day follow-up.
|
Sample Size
|
• 722 cases are expected to be enrolled in this study in China.
|
Study Duration
|
• In this study, it is estimated that the enrollment time will be from January
2021 to June 2023, that is, the enrolment period will be 30 months from
the enrollment of the first subject to the enrollment of the last subject.
After the completion of enrollment, the last subject will be followed up
to 360 days after randomization.
|
2. Statistical Hypothesis
Hypothesis testing will be performed on hematoma recurrence or progression rates.
• Null hypothesis: there is no difference in the incidence of recurrence or progression of
hematoma between the MMA embolization group and the control group at 90 days after
randomization;
• Alternative hypothesis: there is difference in the incidence of recurrence or progression
of hematoma between the MMA embolization group and the control group at 90 days
after randomization.
The above hypothesis tests will be two-sided and statistical significance will be determined
as a p-value of<0.05, and two-sided 95% confidence intervals will be reported.
3. Sample Size Considerations
The study hypothesizes that patients undergoing middle meningeal artery embolization may
have lower symptomatic recurrence (in the trepanation/drainage group) and lower rates of
symptomatic hematoma progression (in the non-surgical group) of subdural hematoma compared
to patients who do not go MMA embolization. Two interim analyses will be conducted. With an
expected symptomatic recurrence rate (trepanation/drainage group) / symptomatic progression
rate (non-surgical group) after MMA embolization of 5% compared with 12% in the control
group, and considering a loss to follow-up rate of 8%, a sample size of 722 cases with a twosided alpha of 0.05 would yield 90% power.
4. Planned Analyses
4.1. Interim Analyses
Two formal interim analyses was planned to review data relating to treatment efficacy,
participant safety and quality of trial conduct using an Haybittle-Peto stopping rule for efficacy.
Given the conservative rule used and the negligible amount of type-I error rate spent at the
interim analysis, the significance threshold will remain at 5% for the final analysis.
4.2. Final Analysis
The planned final analysis will be performed after the following steps are completed
sequentially:
1. All subjects have completed the study as defined in the protocol.
2. The Statistical Analysis Plan (SAP) has been finalized and approved.
3. All required database cleaning is completed, and the head of Data Management department
announces the final database release and database freeze.
4. Randomization codes are issued according to the unblinding process at the end of the trial.
Only blinded data will be reviewed prior to database lock. In addition, no database lock will
occur prior to finalization of this SAP.
5. Analysis Population
Population
|
Definition/Criteria
|
Analyses Evaluated
|
All included
Subjects Set
|
• All subjects who signed informed consent.
|
• Study Population
|
Intent-to-treat
Analysis Set (ITT)
|
• Including all randomized subjects based on
the intention-to-treat (ITT) principle, i.e.
analysis of subjects based on initial
treatment assigned by randomization
|
• Study Population
• Efficacy
|
Per-Protocol
Analysis Set (PPS)
|
• The PPS will include all subjects in the FAS
who did not have any major protocol
deviations.
• Protocol deviations excluding subjects from
the PPS are defined in Section 5.1 (Protocol
Violations).
• Analysis in PPS will be based on the
treatment the participant assigned by
randomization.
• Additional efficacy analyses based on the
PPS set will not be performed if the PPS set
includes 90% or more of subjects in the FAS
and the difference in analysis results
between the two analysis sets is expected to
be negligible.
|
• Study Population
• Efficacy
|
Safety Set (SS)
|
• Includes all randomized subjects.
• As treated
• ITT analysis set
|
• Safety
|
5.1. Protocol Deviations
• Important protocol deviations, including those related to study inclusion/exclusion criteria,
trial conduct, subject management, or subject assessment, will be summarized.
• Protocol deviations will be tracked by the study team during the course of the study
according to relevant regulations in routine monitoring.
− All data will be reviewed prior to database lock to ensure that all important protocol
deviations and deviations that could lead to exclusion of subjects from the analysis can be
captured and classified in the protocol deviation dataset.
− This dataset will serve as the basis for summaries of protocol deviations.
6. Considerations for Data Analyses and Data Handling Conventions
Analyses will be performed using SAS ® System Version 9.4 or higher, and all tables,
figures, and listings will be generated as RTF files.
For descriptive statistics, the number of observations (n), standard deviation, median,
minimum, maximum,Q1 and Q3 will be presented for continuous variables and number and
percentage of subjects will be presented for categorical variables, unless otherwise specified.
Table 2 provides an overview of the appendices in the SAP for outlining general rules for
data analyses and data handling conventions.
Table 2 Overview of Appendix
Component
|
Appendix 1: Treatment Period and Study Period
|
Appendix 2: Data Display Standards and Handling Conventions
|
Appendix 3: Handling of Premature Withdrawals and Missing Data
|
Appendix 4: Abbreviations Table
|
7. Study Population Analyses
7.1. Overview of Planned Study Population Analyses
Unless otherwise stated, the analyses will be based on the ITT set
Table 3 provides an overview of the planned study population analyses.
Table 3 Overview of Planned Study Population Analyses
|
Data Presentation Generated
|
Table
|
Figure
|
Listing
|
Subject Disposition 1, 3
|
Subjects who entered screening and failed screening
|
Y
|
|
Y
|
Randomized Subjects
|
|
Subjects receiving any study treatment
- Received burr hole drainage
- Received MMA embolization
- Received optimal medical therapy
|
|
Completed Subjects
|
|
Subjects who premature withdrew and reasons
|
|
Entered Intent-to-Treat Set (ITT)
|
|
Entered Per-Protocol Analysis Set (PPS)
|
|
Entered Safety Set (SS)
|
|
Protocol Deviations
|
Important protocol deviations
|
Y
|
|
Y
|
Any protocol deviations
|
Y
|
|
Y
|
Demographic and Baseline Characteristics
|
Sex 1
|
Y
|
|
Y
|
Age (years) 2
|
|
Ethnic 1
|
|
Weight (kg) 2
|
|
Height (m) 2
|
|
BMI (kg/m 2) 2
|
|
Smoking status 1
|
|
Alcohol status 1
|
|
Baseline mRS score2
|
|
Prior medical history 1
|
Prior medical history
|
Y
|
|
Y
|
Prior medication history 1
|
Prior antiplatelet, anticoagulant use
|
Y
|
|
Y
|
Prior statin use
|
|
Prior hormonal use
|
|
Baseline cranial imaging diagnosis (CT/MRI)
|
Massive cerebral infarction 1
|
Y
|
|
Y
|
Intracranial space-occupying lesion 1
|
|
Midline shift 1
|
|
If yes, shift distance 2
|
|
Subdural hematoma 1
|
|
If yes, hematoma Side Positions 1
|
|
Maximum thickness of hematoma on the enrolled
side 2
|
|
Volume of hematoma on the enrolled side 2
|
|
Notes:
1. Categorized data.
2. Continuous data.
3. Will be based on all screened sets of subjects.
Y = Plan Generation.
7.2. Description Of Study Population Analysis
7.2.1. Demographic and Baseline Characteristics
• Age (years) will be calculated as (date of informed consent – date of birth)/365.25 rounded
down.
• BMI (kg/m 2) will be calculated as [weight(kg) / (height(cm)/100) ^2].
8. Primary Efficacy Endpoints Analyses
8.1. Overview of Planned Primary Efficacy Endpoints Analyses
Unless otherwise stated, the primary efficacy endpoints analysis will be based on the Intentto-Treat (ITT) and Per-Protocol (PPS) sets. Intent-to-Treat (ITT) set will be the primary analysis
set.
Table 4 provides an overview of the planned primary statistical analyses.
Table 4 Overview of planned primary efficacy endpoints analyses
|
Data Presentation Generated
|
Table
|
Figure
|
Listing
|
Incidence of recurrence (subjects with trepanation) or progression (subjects with no
trepanation) of subdural hematoma within 90 days after randomization
|
Number and percentage of subjects with event
- Hematoma recurrence (in the
trepanation/drainage group) in the MMA
embolization and control arm respectively
- Hematoma progression (in the non-surgical
group) in the MMA embolization and control arm
respectively
- Death within 104 days (90 days + 14 days) in the
MMA embolization and control arm respectively
|
Y
|
|
Y
|
Dichotomous data analyses:
- Recurrence (in the trepanation/drainage group)/
progression rate (in the non-surgical group) with
95% CI
- Difference in recurrence or progression rates
between MMA embolization and control groups
with 95% CI and CMH Chi-square P value
- OR and its 95% CI for the comparison of
recurrence or progression rates between treatment
and control groups
|
|
8.2. Specifications of Primary Efficacy Endpoints Analyses
8.2.1. Definitions and Derivations
• Symptomatic SDH recurrence (in the trepanation/drainage group) refers to a situation in
which the maximum thickness of hematoma of patients exceeds 10 mm, combined with
neurological symptoms, or the need for repeat surgery.
• Symptomatic SDH progression (in the non-trepanation/drainage group) refers to a situation
in which the maximum thickness of increases by more than 3 mm compared with baseline,
combined with neurological symptoms, or the need for surgery.
• The results of central imaging evaluation and analysis of CT/MRI examinations required for
the determination of the subject 's primary endpoint in this study will be presented in tables
and listings, respectively. Central image evaluation will be performed by the independent
Core Lab. Evaluation of neurological symptoms will be derived from the Outcome
Assessment Committee (OAC) review data.
• It should be noted that if there is a record of reoperation in participants who have undergone
trepanation/drainage, and there is a record of surgical treatment in subjects who have
received medical therapy without trepanation/drainage, the occurrence of the primary
endpoint can be directly determined, and hematoma thickness or neurological symptoms are
no longer required.
• In case of non-negligible amounts of missing data (>5%), we will use the MCMC method
based on the Missing at random (MAR) assumption to impute missing data. The regression
model will include group, sex, age, BMI, baseline mRS score and baseline maximum
hematoma thickness. The number of imputations is 10, seed=1234, nbiter=200. SAS code
examples for multiple imputation will be provided in Appendix 3.
• Hematoma recurrence (trepanation/drainage group) or symptomatic hematoma progression
(non-surgical group) at day 90 will be analyzed as follows. If there is any discrepancy
between EDC and central imaging assessment scan date in the determination of visit
window, the date determined by central imaging assessment shall prevail.
Table 5. Data Handling Rules for Primary Endpoint Analysis
Analyses
|
Death within 104
days (90 days + 14
days)
|
Visit out of window
|
No 90-day (± 14
days) visit data
|
Primary
analyses
|
As if an event occurred
|
Observed measurements for
out-of-window visits will be
treated as 90-day results
|
Multiple
imputation*
|
Sensitivity
Analyses1
|
As if an event occurred
|
As No Event
|
As if an event
occurred
|
Sensitivity
Analyses2
|
As if an event occurred
|
As No Event
|
As No Event
|
*If percentage of missing primary endpoint is less than 5%, multiple imputation will not be
performed and missing data will be regarded as no event.
8.2.2. Analysis Methods
• The primary analysis will compare groups using the CMH chi-square test (the stratification
factor is whether or not to perform trepanation/drainage was performed), summarize the 90-
day recurrence (trepanation/drainage group) / progression rate (non-surgical group) for the
MMA embolization group and the control group using the two-sided 95% CI (ClopperPearson method) and summarize the two-sided 95% CI for the difference in recurrence or
progression rates between the two groups using the Miettinen-Nurminen method.
• Sensitivity analyses will use generalized linear mixed-effects model to calculate ORs and their
95% CIs for the comparison of recurrence or progression rates between the two arms, with
group as fixed effect, site as random effect. Results of multivariable analysis will also be
presented, including treatment group, sex, age, surgical treatment strata (trepanation/drainage
vs. no trepanation/drainage), any usage of antiplatelet or anticoagulant therapy, hematoma
volume of baseline CT as covariates.
8.2.3. Model Specifications
8.2.3.1. Model Specifications of Primary Analysis
Statistical analysis of binary categorical variables
|
Endpoints and parameters
|
• Endpoint: recurrence or progression rate of hematoma at 90 days after randomization
• Parameters:
− Proportion estimate and 95% CI
− Risk difference, p-value and 95% CI
|
Input Variables
|
• AVAL: response variable
• TRTAN: groups
• COUNT: number of subjects per level per group
• COVARIATES: adjustment Variables
|
SAS Code Sample
|
Example code for Clopper-Pearson 95% CI calculation:
proc freq data = adeff;
table aval/binomial (cl = exact) alpha = 0.05 ;
weight count;
by trtan;
ods output binomialcls = binomialcl;
run;
Example code for risk difference and 95% CI calculation:
proc freq data = adeff;
table trtanaval/riskdiff (cl = mn) chisq alpha = 0.05;
weight count;
ods output pdiffcls = diff chisq = pvalue1;
run;
Example code for CMH chi-square:
proc freq data = adeff;
table stratatrtanaval/chisq alpha = 0.05;
weight count;
ods output chisq = pvalue1;
run;
|
Presentation of Results
|
• Estimates of rates and 95% CIs were available for BinomialCL.
• Risk difference and 95% CI are available from diff.
• Chi-square P values are available from PVALUE1.
|
8.2.3.2. Model Specifications of Sensitivity Analysis
Statistical analysis of binary categorical variables
|
Endpoints and parameters
|
• Endpoint: hematoma recurrence (trepanation/drainage group) or symptomatic hematoma
progression (non-surgical group) rate of hematoma at 90 days after randomization
• Parameters:
− OR and 95% CI
|
Input Variables
|
• AVAL: response variable
• TRTAN: groups
• SITEID: subject site number
• COUNT: number of subjects per level per group
• COVARIATES: adjustment Variables
|
SAS Code Sample
|
Generalized Linear Mixed-effects Model Example Code:
proc glimmix data=adeff descending;
class site trtan;
model aval/count = trtan covariates / solution;
random intercept / subject=site;
ods output oddsratio= odds ratio estimates;
run;
|
Presentation of Results
|
• The OR and 95% CI for the ratio can be obtained from Odds Ratio Estimates.
|
8.3. Subgroup Analyses
The primary efficacy endpoint point will be analyzed in the following subgroups:
• Age group(<70 vs ≥70)
• Sex (male vs female)
• Surgical treatment strata (trepanation/drainage vs. no trepanation/drainage)
• Smoking status(yes vs no)
• Medical history of brain trauma (yes vs no)
• Any usage of antiplatelet or anticoagulant therapy before randomization(yes vs no)
• Any usage of statins within one month before randomization(yes vs no)
• Midline shift of baseline CT (<10 mm vs ≥10 mm)
• Hematoma thickness of baseline CT (<10 mm vs ≥10 mm)
• Hematoma volume of baseline CT (<Q3 of combined hematoma volume data, ≥Q3
of combined hematoma volume data)
• Bilateral SDH (yes vs no)
• Embolized branches of MMA (main trunk only, distal only, main trunk & distal)
Subgroup analysis will use the same statistical analysis methods as described in Section 8.2.2.
9. Secondary Statistical Analyses
9.1. Secondary Efficacy Endpoint Analyses
9.1.1. Overview of Planned Analyses for Secondary Efficacy Endpoints
Unless otherwise stated, analyses of secondary efficacy endpoints will be based on the
intent-to-treat analysis set (ITT) and per-protocol (PPS) set.
Table 6 Planned Overview of Secondary Efficacy Endpoint Analyses
|
Data Presentation Generated
|
Table
|
Figure
|
Listing
|
Incidence of recurrence or progression of SDH at 360 days after randomization:
as Table 4 Overview of planned primary statistical analyses
|
Categorical secondary endpoints:
Success rate of target blood vessel embolization in DSA imaging
Change in mRS score at 90 days and 360 days after randomization compared to baseline
Proportion of patients with mRS score of 0 to 2 at 90 days and 360 days after randomization
Proportion of patients with mRS score of 0 to 3 at 90 days and 360 days after randomization
Destination for rehabilitation after discharge (going home vs rehabilitation hospital)
|
Statistical description of categorical data:
Number (n) and percentage of subjects in each
category
|
Y
|
|
Y
|
Dichotomous data analysis:
- Proportion and its 95% CI
- Difference in sample rate between MMA
embolization and control groups, p-value and 95%
CI
- OR for MMA embolization and control groups
with 95% CI
|
|
Continuous Secondary Endpoints:
Changes of hematoma thickness in CT or MRI imaging at 90 days after randomization
compared to baseline
Changes of hematoma volume at 90 days after randomization
Changes of midline shift in CT or MRI imaging at 90 days after randomization
Changes of mRS score at 90 days and 360 days after randomization
Quality of life evaluated with EQ-5D-5L Scale at 90 days and 360 days after randomization
Total hospital stay (number of days)
Re-hospitalization times
Total hospitalization expenses
|
Statistical description of continuous data:
- Number of non-missing cases (n)
- Mean (standard deviation)
- Median
- Min, Max
- Q1,Q3
|
Y
|
|
Y
|
Continuous data analysis:
- Estimate of mean and 95% CI for MMA
embolization and control groups
- Difference in means and 95% CI between MMA
embolization and control groups
- P value for difference in means between MMA
embolization and control groups
|
|
9.1.2. Specifications of Secondary Efficacy Endpoint Analyses
9.1.2.1. Definitions and Derivations
• Change from 90 days/360 days post study treatment will be calculated as 90 days/360 days
measurement – baseline measurement
• Total days in hospital = Date of discharge - Date of hospitalization + 1 (Hospital information
page)
• Other definitions and derivations are the same as in Section 8 .2.1
9.1.2.2. Statistical Analysis Methods
• 360 days recurrence or progression of SDH after randomization will be analyzed in the same
way as the primary endpoint (Section8.2.2)
• Ordinal logistic regression will be used for intergroup comparison of mRS scale scores at 90
and 360 days after randomization, with treatment group, sex, age, surgical treatment strata
(trepanation/drainage vs. no trepanation/drainage), any usage of antiplatelet or anticoagulant
therapy within one month before randomization(yes vs no), hematoma volume of baseline
CT as covariates.
• Other categorical secondary endpoints will be analyzed in the same way as the primary
endpoint (Error! Reference source not found.Error! Reference source not
found.Section)
• The linear mixed effects model approach will be used to estimate the mean estimate (95%
CI) and the mean estimate (95% CI) of the differences between groups as well as the P
values of the differences between groups for the change in maximum hematoma thickness at
90 days after randomization compared to baseline and the change in hematoma volume at 90
days after randomization compared to baseline; Treatment group variables, sex, age, surgical
treatment strata (trepanation/drainage vs. no trepanation/drainage), any usage of antiplatelet
or anticoagulant therapy within one month before randomization(yes vs no), hematoma
volume of baseline CT , visit variables, and visit by treatment group variable interactions
will be included in the model and site as the random effect.
• For other continuous secondary endpoints, T tests will be used to compare groups, and 95%
confidence intervals for the difference in means between groups will be calculated using
normal approximation.
9.1.2.3. Model Specifications
Statistical analysis of binary categorical variables
|
Endpoints and parameters
|
• Endpoint: Change of mRS scale score at 90 and 360 days after randomization compared to
baseline
• Parameters:
− Ordinal OR and 95% CI
|
Input Variables
|
• AVAL: response variable
• TRTAN: groups
• SITEID: subject site number
• COUNT: number of subjects per level per group
• COVARIATES: adjusted Variables
|
SAS Code Sample
|
Logistic Regression Example Code:
proc glimmix data=adeff descending;
class siteid trtan;
model aval/ count = trtan covariates / solution;
random intercept / subject= siteid;
run;
|
Presentation of Results
|
• The OR and 95% CI for the ratio can be obtained from Odds Ratio Estimates.
|
Statistical analysis of continuous variables
|
Endpoints and parameters
|
• Endpoint:
Changes of hematoma thickness in CT or MRI imaging at 90 days after randomization
compared to baseline
Changes of hematoma volume at 90 days after randomization compared to baseline
Changes of midline shift in CT or MRI imaging at 90 days after randomization compared
to baseline
Quality of life evaluated with EQ-5D-5L Scale at 90 days and 360 days after
randomization compared to baseline
Total hospital stay (number of days)
Re-hospitalization times
Total hospitalization expenses
• Parameters:
− Mean and 95% CI
− Difference in means and 95% CI with P-value
|
Input Variables
|
• AVAL: response variable
• TRTAN: groups
• SITEID: subject site number
• COUNT: number of subjects per level per group
• COVARIATES: adjusted Variables
|
SAS Code Sample
|
Linear Mixed Effects Model Method Example Code:
proc mixed data = adeff;
class trtan siteid avisitn;
model aval = trtan covariates avisitn avisitn avisitn trtan/solution ddfm = kr;
random siteid /type = cs;
lsmeans trtanavisitn/diff cl alpha = 0.05 e;
ods output lsmeans = lsmeans diffs = diffs;
run;
T-Test Example Code:
ods otput conflimits = conflimits ttests = ttests;
proc ttest data = adeff dist = normal sides = 2 ci = equal alpha = 0.05;
class trtan;
var aval;
run;
|
Presentation of Results
|
Linear mixed effects model approach:
• Estimation of mean and 95% CI for both groups:
o Point Estimate: Estimate Variable in lsmeans dataset
o Corresponding 95% CI: Lower vs. Upper variables in lsmeans dataset
• Estimate and 95% CI for difference in means between groups:
o Filtered all TRT A N ≠ TRT A N and AVISITN = AVISITN records in Diffs dataset
and output:
- Point Estimate: Estimate Variable in Diffs Post-Screening Dataset
- Corresponding 95% CI: Lower versus Upper Variables in Diffs Post-Screening
Dataset
• P-value: Probt variable in filtered dataset Diffs
T-Test:
• The difference in means and 95% CI are available from CONFLIMITS.
• P-values for t-tests are available from TTESTS.
|
9.2 Safety analyses
9.2.1 Overview of Planned Safety Analyses
Unless otherwise stated, safety analyses will be based on the Safety Analysis Set.
Table 7 provides an overview of the planned safety analyses.
|
Data Presentation Generated
|
Table
|
Figure
|
Listing
|
Adverse events (within 90 days/360 days after surgery) 1
|
All Adverse Events
|
Y
|
|
Y
|
Serious Adverse Events (SAEs)
|
|
Moderate and severe adverse events
|
|
Procedure-related serious complications within 30 days after randomization (including
surgical and interventional procedures) 1, 2
|
Major complications associated with drilling or
MMA embolization
|
Y
|
|
Y
|
Major complications associated with drilling
|
|
Major complications associated with MMA
embolization
|
|
Adverse events of special interest 1
|
Adverse events of special interest
|
Y
|
|
Y
|
Serious adverse events of special interest
|
|
Death (within 90 days/360 days after randomization) 1
|
Death from any cause
|
Y
|
|
Y
|
Concomitant medications 1
|
Antiplatelet or anticoagulant therapy
|
Y
|
|
Y
|
Other concomitant medications
|
|
Notes:
1. Categorical data.
2. Postoperative serious complications include: symptomatic intracranial hemorrhage, surgery-related
intracranial hemorrhage, surgery-related neurological dysfunction , surgery-related central nervous system
infection , surgery-related arterial dissection, vessel wall injury, and vessel rupture and perforation , surgeryrelated ischemic events , retroperitoneal/wrist hematoma , puncture site neuropathy , contrast agent allergy, or
contrast agent encephalopathy
Y = Plan Generation.
9.2.2 Specifications Of Safety Analyses
9.2.2.1 Adverse Events
• An untoward medical occurrence during a clinical investigation, whether or not related to
the investigational medical device, is an adverse event (AE).
• AEs will be coded using MedDRA version 24.0 or higher. System Organ Class (SOC) and
Preferred Term (PT) will be reported.
• The investigator will grade the severity of the AE (mild, moderate, severe). AEs with
missing severity will not be imputed.
• Adverse events will be summarized using frequency tables in descending order of incidence
by SOC and PT unless otherwise specified. If a subject has the same AE reported more than
once, the highest severity of this AE for that subject will be reported.
• For adverse events of special interest, the CEC assessment and investigator assessment will
be performed, and the results of the two assessments will be presented separately.
9.2.2.3 Concomitant medication
• Concomitant medications are defined as medications that were taken by the patient until or
until after randomization.
• Concomitant medications will be summarized by treatment group and the number and
percentage of subjects will be listed by medication name.
10. References
[1]. Zuo, Q., Ni, W., Yang, P., Gu, Y., Yu, Y., Yang, H., ... & MAGIC-MT investigators.
(2023). Managing non-acute subdural hematoma using liquid materials: a Chinese
randomized trial of middle meningeal artery treatment (MAGIC-MT)—protocol. Trials,
24(1), 586.
[2]. Newcombe, R. G. (1998). Two‐sided confidence intervals for the single proportion:
comparison of seven methods. Statistics in medicine, 17(8), 857-872.
[3]. Newcombe, R. G. (1998). Interval estimation for the difference between independent
proportions: comparison of eleven methods. Statistics in medicine, 17(8), 873-890.
[4]. Rayner, J. C. W., & Rippon, P. (2018). Recent extensions to the cochran–mantel–haenszel
tests. Stats, 1(1), 98-111.
[5]. Jiang, J., & Nguyen, T. (2007). Linear and generalized linear mixed models and their
applications (Vol. 1). New York: Springer.
[6]. Bender, R., & Grouven, U. (1997). Ordinal logistic regression in medical research. Journal
of the Royal College of physicians of London, 31(5), 546.
[7]. Lewis, J. A. (1999). Statistical principles for clinical trials (ICH E9): an introductory note on
an international guideline. Statistics in medicine, 18(15), 1903-1942.
[8]. Jennison, C., & Turnbull, B. W. (1999). Group sequential methods with applications to
clinical trials. CRC Press.
12. APPENDICES
12.1 Appendix 1: Treatment Period and Study Period
Treatment Period and Study Period
|
Study Day
|
Day 0 is defined as the day of Randomization.
Study duration after Day 0 is calculated as the number of days relative to randomization date:
• Reference Date = Missing Value → Study Day = Missing Value
• Reference Date < Randomization → Study Day = Reference Date – Randomization Date
• Reference Date ≥ Randomization → Study Day = Reference Date – randomization Date +
1
|
Time points and assessment windows
|
Study Flow Chart
|
STUDY
PROCEDURES
|
Screeninga
|
Baseline
|
Follow-up Period
|
Visit Timeb
|
V0
|
V1
|
V2
|
V3
|
V4
|
V5
|
-14 to Day 0
|
Day 0
|
At discharge
|
Day 30
|
Day 90
|
Day 360
|
Informed Consent
|
X
|
|
|
|
|
|
Demographics
|
X
|
|
|
|
|
|
Inclusion/Exclusion Criteria
|
|
X
|
|
|
|
|
Medical History and Prior Medications
|
X
|
|
|
|
|
|
Vital signs
|
X
|
|
|
X
|
X
|
X
|
Admission examinationc
|
X
|
|
|
|
|
|
CT/MRId
|
X
|
|
|
|
X
|
X
|
Informed Consent
|
X
|
|
|
|
|
|
Demographics
|
X
|
|
|
|
|
|
Inclusion/Exclusion Criteria
|
|
X
|
|
|
|
|
Medical History and Prior Medications
|
X
|
|
|
|
|
|
Vital signs
|
X
|
|
|
X
|
X
|
X
|
Treatment Period and Study Period
|
Admission examination c
|
X
|
|
|
|
|
|
CT/MRI d
|
X
|
|
|
|
X
|
X
|
DSAc
|
|
X
|
|
|
|
|
Randomized and treated
|
|
X
|
|
|
|
|
Treatment and nursing costs
|
|
|
X
|
|
X
|
X
|
Efficacy Endpoints
|
|
|
|
|
X
|
X
|
Serious Adverse Events
|
|
|
X
|
SDH-related death events
|
|
|
Procedure-related serious complications
|
|
|
Concomitant medication/treatment
|
|
|
X
|
X
|
X
|
X
|
MRS scale
|
X
|
|
|
|
X
|
X
|
EQ-5D Scale
|
X
|
|
|
|
X
|
X
|
|
A Screening visit and baseline visit may be on the same day
B V3 visit window is ±7 days, V4 visit window is ± 14 days, and V5 visit window is ± 30 days
C Blood routine (red blood cell count, white blood cell count, platelet count, hemoglobin), coagulation
function (APTT ,INR), liver and kidney function (ALT , AST ,BUN ,Cr), Electrolytes (potassium, sodium,
chloride, calcium), fasting blood glucose; Women of childbearing potential will need a negative urine HCG
test to be enrolled. Those women of childbearing potential with positive or missing pregnancy test will not
be included.
D CT is mandatory, MRI optional, and MRI examination may be performed at the physician 's discretion
according to the patient' s condition
E Performed only in the MMA embolization group
|
Treatment Period and Study Period
|
Time Point
|
Acceptable Time Window
|
V3
|
± 7 days
|
V4
|
± 14 days
|
V5
|
± 30 days
|
|
12.2 Appendix 2: Data Display Standards and Handling Conventions
Data Display Standards and Handling Conventions
|
Criteria for Data Presentation
|
• Numerical data will be reported with precision collected in the eCRF.
• Precision reported from non-eCRF sources will follow dMed 's statistical principles but
may be adjusted to clinically interpretable decimal places (dp.).
• Derived data and their statistics will be 1 decimal place more than the original data and
the corresponding statistics. For example, if the raw data collected contains 1 decimal
place, derived data in the listings will be presented to 2 decimal places. Therefore, the
average number of derived data will be presented with 3 decimal places, minimum and
maximum with 2 decimal places, standard deviation with 4 decimal places, and so on.
• Precision validity and display utility will be considered in determining the number of
decimal places displayed. Therefore, if the number of decimal places is too large to add
additional information, the reported data will retain the appropriate number of decimal
places. The maximum number of decimal places reported for any summary statistic should
be 4, unless otherwise specified.
• The maximum number of decimal places can be directly applied to the ADaM dataset.
|
Data Display Standards and Handling Conventions
|
Precision
|
Name
|
Description
|
Decimal place (dp)
|
N
|
Number of subjects in treatment group
|
Always shown as 0 dp.
|
N
|
Number of subjects with non-missing results
|
Always shown as 0 dp.
|
%
|
Percent
|
Categorical data shown as 1 dp.
|
Mean
|
Arithmetic mean
|
1 dp. more than original data
|
SD
|
Standard deviation
|
2 dp. more than original data
|
Median
|
Median
|
1 dp. more than original data
|
Min.
|
Minimum value
|
Corresponds to raw data
|
Max.
|
Max
|
Corresponds to raw data
|
SE
|
Standard Error
|
1 dp. more than statistical parameters
|
95% CI
|
95% confidence interval
|
1 dp. more than statistical parameters
|
P value
|
P value
|
3 dp., or < 0.001
|
|
Baseline Definitions and Derivations
|
The last non-missing measurement/assessment prior to the randomization date will be used as
the baseline measurement unless specified.
Measurements/assessments will be considered (if not missing) at baseline if performed on the
same day as randomization.
• Change from baseline = value at post-treatment visit – baseline if there is no specific formula
or medical requirement in the protocol.
• Percent change from baseline = 100% * (treatment value at post visit – baseline)/baseline
|
General Data Display Standards and Handling Conversion
|
Data Display Standards and Handling Conventions
|
Planned and Actual Time
• Unscheduled visits and visits outside protocol-specified windows (ie, documented as
protocol deviations) will be included in listings only, unless otherwise specified.
Proportion Calculation
• Percent of zero counts will not be shown.
• Percentages will be calculated based on the number of participants in the analysis
population of interest, unless specifically stated. Missing observations will also be
included in the denominator calculation.
Time Transition of segments
• 1 year = 365.25 days
• 1 month = 30.4375 days
• 1 week = 7 days
|
12.3 Appendix 3: Handling of Early Withdrawals and Missing Data
Handling of Early Withdrawals and Missing Data
|
Subjects Withdrawn Early
|
Subject substitution: Subjects will not be replaced regardless of withdrawal from the trial for
any reason.
|
Handling of missing data
|
For the analysis of the primary endpoint, no 90 day (± 14 days) visit data subjects will be
handled as described in the table below.
|
Analysis
|
Death within 104 days
(90 days + 14 days)
|
Visit out of window
|
No 90-day (± 14
days) visit data
|
Handling of Early Withdrawals and Missing Data
|
Primary
analysis
|
As if an event occurred
|
Observed measurements for
out-of-window visits will be
treated as 90-day results
|
Multiple
imputation*
|
Sensitivity 1
|
As if an event occurred
|
As No Event
|
As if an event
occurred
|
Sensitivity
Analysis 2
|
As if an event occurred
|
As No Event
|
As No Event
|
*If percentage of missing primary endpoint is less than 5%, multiple imputation will not be
performed and missing data will be regarded as no event.
SAS Code Sample:
proc mi data=im1 seed=1234 nimpute=20 out=im2
round=. . . . . 1
min=. . . . . 0
max=. . . . . 1;
;
by trtan;
var sex age bmi basemrs basemth aval;
mcmc chain=multiple nbiter=200 niter=100;
run;
proc sort data=im2;
by _imputation_ trtan usubjid siteid;
run;
proc transpose data=im2 out=im2_t(rename=(_name_=avisit col1=aval));
by _imputation_ trtan usubjid siteid;
var v1 v2;
run;
proc sql noprint;
create table im2_count as
select distinct
_imputation_,
trtan,
avisit,
aval,
count(*) as count
from im2_t
group by _imputation_,trtan,avisit,aval
order by _imputation_,trtan,avisit,aval
;
quit;
/*clopper-pearson 95%ci*/
ods select none;
proc freq data= im2_count;
by _imputation_ trtan avisit;
table aval / binomial (cl=exact) alpha=0.05;
weight count;
ods output binomial=binomial;
run;
ods select all;
proc transpose data=binomial out=binomial_t;
by _imputation_ trtan avisit;
var nvalue1;
id label1;
quit;
proc sort data=binomial_t;
by trtan avisit _imputation_;
run;
ods select none;
proc mianalyze data=binomial_t;
by trtan avisit;
modeleffects proportion;
stderr ase;
ods output parameterestimates=binomial_t_pool;
run;
ods select all;
/*mn*/
proc freq data=im2_t;
by _imputation_ ;
tables trtan *aval / commonriskdiff (cl=score test=score) riskdiff (cl=mn);
ods output commonpdifftests=commonpdifftests1 commonpdiff=commonpdiff1
pdiffcls=pdiffcls1 riskdiffcol1=riskdiffcol11;
run;
data riskdiffcol11;
set riskdiffcol11(where=(row="difference"));
run;
proc sort data=riskdiffcol11;
by _imputation_;
run;
proc mianalyze data=riskdiffcol11;
modeleffects risk;
stderr ase;
ods output parameterestimates=pest1_pool;
run;
/*cmh*/
*** perform cmh test;
proc freq data=im2_t;
tables strata*trtan*aval/cmh;
ods output cmh=cmh;
by _imputation_;
run;
*** apply wilson-hilferty transformation to the cmh statistic and
standardize the resulting normal variable;
data cmh_wh;
set cmh(where=(althypothesis="general association"));
cmh_value_wh=((value/df)**(1/3) - (1-2/(9*df)))/sqrt(2/(9*df));
cmh_sterr_wh = 1.0;
run;
*** combine results;
proc mianalyze data=cmh_wh;
ods output parameterestimates=mian_cmh_wh;
modeleffects cmh_value_wh;
stderr cmh_sterr_wh;
run;
proc sql noprint;
create table im2_count2 as
select distinct
_imputation_,avisit,siteid,trtan,aval,
count(*) as count
from im2_t
group by _imputation_,avisit,siteid,trtan,aval
order by _imputation_,avisit,siteid,trtan,aval
;
quit;
ods select none;
proc glimmix data=im2_count2;
by _imputation_ avisit;
class siteid trtan aval;
model aval/count=trtan / solution;
random intercept / subject=siteid;
ods output parameterestimates=parameterestimates;
run;
ods select all;
proc sort data=parameterestimates(where=(trtan=1)) out=parameterestimates1;
by trtan avisit _imputation_ ;
run;
ods select none;
proc mianalyze data=parameterestimates1;
by trtan avisit;
modeleffects estimate;
stderr stderr;
ods output parameterestimates=parameterestimates1_pool;
run;
ods select all;
Prior/concomitant medications/treatments
Prior medications/therapies are those that ended prior to randomization.
Concomitant medications/therapies are medications/therapies other than study treatment or
premedication with study treatment taken at any time during the study (on or after
randomization), including medications/therapies started and continued prior to randomization.
Incomplete dates for prior/concomitant/post-study medications/treatments recorded in the CRF
will not be imputed and will be classified according to the following rules:
Date Started
|
End date
|
Actions
|
Not missing/partially missing/missing
|
Not missing
|
• Prior medication/therapy if end date < randomization date
• Concomitant medication/treatment if end date ≥ randomization date
|
Partially missing, but known to partially show that it cannot occur on or after the date of randomization
|
• Prior Medications/Therapies
|
Partial missing, unclear
relationship to randomization
date
|
• Assumed concomitant
medication/treatment
|
Complete absence
|
• Assumed concomitant
medication/treatment
|
Note: "<" refers to "earlier than" and "≥" refers to "same day or later".
12.4 Appendix 4: Abbreviations Table
Abbreviation
|
Description
|
AE
|
Adverse Event
|
CRF
|
Case Report Form
|
CT
|
Computed Tomography
|
DSA
|
Digital Subtraction Angiography
|
EQ-5D-5L
|
A standardized assessment instrument (developed by the EuroQolGroup) that
provides a simple descriptive measure of health-related quality of life
|
MMA
|
Middle Meningeal Artery
|
MRI
|
Magnetic Resonance Imaging
|
MRS
|
Modified Rankin Scale
|
PT
|
Preferred Term
|
RTF
|
Rich Text Format
|
SAE
|
Serious Adverse Event
|
SAP
|
Statistical Analysis Plan
|
SDH
|
Subdural Hematoma
|
SOC
|
System Organ Class
|
|
|