Skip to main content
Erschienen in: Trials 1/2019

Open Access 01.12.2019 | Study protocol

The effectiveness and safety of Kami Guibi-tang for mild cognitive impairment: study protocol of a pilot, randomized, placebo-controlled, double-blind trial

verfasst von: Hee-Yeon Shin, Jeong-Hwa Kim, Geon-Ho Jahng, Woo-Sang Jung, Seong-Uk Park, Chang-Nam Ko, Jung-Mi Park

Erschienen in: Trials | Ausgabe 1/2019

Abstract

Background

Mild cognitive impairment (MCI) is an intermediate phase between normal aging and dementia. Since a majority of amnestic MCI (aMCI) cases progress to Alzheimer’s disease (AD), it is considered the prodromal stage of AD and, therefore, a treatment target for the prevention of further cognitive decline. However, there is no approved treatment for MCI at present. Kami Guibi-tang (KGT) is a herbal drug used in Korean medicine to treat amnesia, insomnia, loss of appetite, and depression. We will explore the effectiveness and safety of KGT in amnestic MCI in this trial.

Methods/design

The study will be a single-center, randomized, placebo-controlled, double-blind trial. Eligible participants diagnosed with amnestic MCI will be randomly allocated to a treatment or control group. Participants will take KGT or placebo granules, three times a day, for 24 weeks. The primary outcomes will be changes in Seoul Neuropsychological Screening Battery (SNSB) scores, and magnetic resonance imaging (MRI) measurements including those of brain metabolites, neurotransmitters, and cerebral blood flow. The secondary outcomes will include the safety assessment, measured by changes in blood chemistry, changes in blood protein and cholesterol levels related to AD pathology, and a comparison of MRI changes between the two groups, using age and genotype as covariates.

Discussion

This study will be the first clinical trial to identify the therapeutic potential of Kami Guibi-tang for amnestic MCI. The findings will provide insight into the feasibility of large-scale trials to gather evidence for KGT as a treatment for MCI.

Trial registration

Korean Clinical Trial Registry, ID: KCT0002407. Registered on 30 March 2017.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13063-019-3567-1) contains supplementary material, which is available to authorized users.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AD
Alzheimer’s disease
ADAS-cog
Alzheimer’s Disease Assessment Scale-cognitive subscale
ALT
Alanine aminotransferase
aMCI
Amnestic mild cognitive impairment
ASL MRI
Arterial spin-labeling perfusion magnetic resonance imaging
AST
Aspartate aminotransferase
BUN
Blood urea nitrogen
CBF
Cerebral blood flow
CDR
Clinical Dementia Rating
GDS
Global Deterioration Scale
KDSQ
Korean Dementia Screening Questionnaire
KGT
Kami Guibi-tang
K-MMSE
Korean Version of Mini-Mental State Examination
MCI
Mild cognitive impairment
MRS
Magnetic resonance spectroscopy
NINCDS-ADRDA
National Institute of Neurological and Communicative Disorders and Stroke and Alzheimer’s Disease and Related Disorders Association
pCASL MRI
Pseudo-continuous arterial spin-labeling perfusion magnetic resonance imaging
rCBF
Regional cerebral blood flow
SNSB
Seoul Neuropsychological Screening Battery

Background

Mild cognitive impairment (MCI) is an intermediate phase between normal aging and dementia. This clinical condition is characterized by self or caregiver-reported memory or cognitive complaints, and objective cognitive impairment, that is not severe enough to interfere with daily activities [1]. The majority of people with MCI develop dementia due to Alzheimer’s disease (AD) [2]. In particular, the amnestic subtype of MCI, which manifests as memory complaints, is often caused by degenerative etiologies and is generally regarded as a precursor of AD. A previous study revealed that 16% of amnestic MCI (aMCI) patients progress to dementia each year, 99% of whom receive an AD diagnosis [3]. Early treatment of mild-to-moderate AD is associated with better responses than later treatment, so treatment of MCI may delay progression to AD [4]. There is, however, no treatment approved for enhancing memory function, or preventing further cognitive decline in MCI [5].
Kami Guibi-tang (KGT) is a popular herbal medicine used in Korean and Kampo medicine to treat amnesia, insomnia, loss of appetite, and depression. Several studies have explored the effects of KGT on cognitive impairment, particularly in AD. KGT improved learning performance in a mouse model of accelerated senescence [6], and reduced spatial memory impairment induced by scopolamine or delta-tetrahydrocannabinol [7]. In the 5XFAD mouse model of AD, KGT improved deficits in object recognition memory, and reversed the degeneration of cortical axons and presynaptic terminals [8]. A clinical study demonstrated that the Mini-Mental State Examination (MMSE) score of AD patients improved after receiving orally administered KGT extract granules for 3 months [9].
Previous studies have suggested that KGT could be beneficial for cognitive function in AD patients. However, few clinical trials have been published, and no study has yet investigated the effect of KGT on MCI. This pilot study aims to determine the effectiveness and safety of KGT for improving cognitive function, the influence on neuroimaging and neurochemical biomarkers, and the potential therapeutic effects in MCI.

Methods/design

Study design and setting

This study will use a single-center, randomized, double-blind, placebo-controlled, parallel-group clinical trial design. The study will take place at Kyung Hee University Hospital at Gangdong, Seoul, Korea from March 2017 through November 2019. The flow chart of the study design is shown in Fig. 1. The schedules for enrollment, interventions, and assessments are shown in Table 1.
Table 1
The schedule of enrollment, interventions, and assessments (SPIRIT 2013 Statement)
https://static-content.springer.com/image/art%3A10.1186%2Fs13063-019-3567-1/MediaObjects/13063_2019_3567_Tab1_HTML.png

Participants

Inclusion criteria

Participants who meet all of the following criteria will be eligible to participate:
1.
Aged 55–90 years, with complaint of impaired memory
 
2.
Objective cognitive impairment as measured by the Seoul Neuropsychological Screening Battery (SNSB), with a score of 3 on the Global Deterioration Scale (GDS), 0.5 on the Clinical Dementia Rating (CDR), and a normal score on the Korean MMSE (K-MMSE)
 
3.
Diagnosed with aMCI by a neurologist
 
4.
Participants who have not taken medication affecting cognitive function, including gliatilin, gliatamine, ginexin, tanamin, or other psychoactive drugs, in the previous 2 weeks
 
5.
Participants who have not changed medication for underlying diseases in the previous 2 weeks, and no expected change in medication during the study period
 
6.
No difficulty in communicating
 

Exclusion criteria

Participants who meet any of the following criteria will be ineligible to participate:
1.
Diagnosed with AD, based on the criteria of National Institute of Neurological and Communicative Disorders and Stroke and Alzheimer’s Disease and Related Disorders Association (NINCDS-ADRDA)
 
2.
Brain disorders causing neurological symptoms, other than cognitive impairment
 
3.
Parkinson’s disease, Huntington’s disease, Down’s syndrome, Creutzfeldt-Jakob disease, or any other neurodegenerative disorder
 
4.
Cognitive impairment resulting from other diseases including head trauma, hypoxic brain damage, vitamin deficiency, brain tumor, encephalitis, neurosyphilis, and mental retardation
 
5.
Cerebrovascular diseases with magnetic resonance imaging (MRI) evidence
 
6.
A previous or current history of major depression
 
7.
Concomitant psychiatric disorders or behavioral problems that require antipsychotic medication
 
8.
A history of a convulsive disorder, except for febrile convulsion during childhood
 
9.
Unstable or life-threatening medical conditions
 
10.
Uncontrolled hypertension
 
11.
Heart or renal diseases
 
12.
Peripheral edema
 
13.
Gastrointestinal symptoms such as anorexia, nausea, abdominal pain, or diarrhea
 
14.
Taking medication that could induce hypokalemia or myopathy
 
15.
Drug hypersensitivity to the constituents of the study medication
 
16.
Possibility of pregnancy
 
17.
Clinically significant abnormalities in blood chemistry test results, including levels of serum aspartate aminotransferase/alanine aminotransferase (AST/ALT) more than two-fold the upper normal limit, or serum creatinine (Cr) level more than 10% of the upper normal limit
 
18.
Participation in any other clinical trials within the previous 4 weeks
 
19.
Illiteracy
 
20.
Contraindications for MRI
 
21.
Considered unsuitable for participation by the investigators
 

Drop-out criteria

1.
Occurrence of any severe adverse effects
 
2.
Participants’ voluntary withdrawal from the trial
 
3.
Participants not following the protocol (i.e., drug compliance below 80%)
 
4.
Use of additional medication to improve cognitive function during the study period
 
5.
Decision made by the principal investigator
 

Recruitment, enrollment

We will recruit a total of 38 patients through advertisements and referrals and screen the candidates aged 55–90 years, with complaints of impaired memory, using the inclusion/exclusion criteria, the K-MMSE, and the Korean Dementia Screening Questionnaire (KDSQ). Potentially eligible participants will undergo the SNSB, and those who are diagnosed with aMCI by a neurologist will be included in the trial. The investigators will provide a detailed explanation of the purpose, procedures, and potential risks and benefits of the study. Participants willing to participate in the trial and provide biological specimens will sign a consent form prior to enrollment.

Randomization, allocation, blinding

A researcher uninvolved in the assessment will generate a random sequence using SPSS ver. 18. We will randomly allocate participants to either the treatment or the control group, in a 1:1 ratio using the block randomization method, with a block size of 4. The participants, assessor, clinical trial pharmacist, and researchers will be blinded to the allocations throughout the course of the study. Cases will be unblinded only if serious adverse events (SAE) occur.

Intervention

After randomization, the treatment group will receive KGT granules (3 g/pack) and the control group will receive placebo granules (3 g/pack). Kami Guibi-tang, the herbal medicine under study, is composed of 14 drugs: Astragali Radix (1.0 g), Ginseng Radix (1.0 g), Atractylodis Rhizoma (1.0 g), Poria cocos (1.0 g), Zizyphi Fructus (0.67 g), Zingiberis Rhizoma (0.33 g), Saussureae Radix (0.33 g), Glycyrrhizae Radix (0.33 g), Zizyphi Spinosi Semen (1.0 g), Longan Arillus (1.0 g), Angelicae Radix (0.67 g), Polygalae Radix (0.67 g), Bupleuri Radix (1.0 g), Moutan Radicis Cortex (0.67 g), and Gardeniae Fructus (0.67 g). Treatment granules will be manufactured by Kyoung Bang Pharmaceutical Co., Ltd. (Incheon, Korea), which has been certified for Good Manufacturing Practice.
Placebo granules will be produced by the same manufacturer, using the standard method of placebo manufacturing according to the Korean Good Manufacturing Practice guidelines. The placebo will be similar to the Kami Guibi-tang granules in appearance, taste, and smell.
An independent clinical pharmacist will distribute KGT or placebo granules to the study participants. The pharmacist will instruct participants to dissolve the granules in hot water, stir well, and drink the solution three times per day, 30 min after meals, for 24 consecutive weeks. The researchers will call participants by phone once a week, reminding them to comply with the medication schedule.
The administration of medicine for underlying diseases, such as hypertension or diabetes mellitus, will be permitted during the intervention; however, any medication that can affect cognitive function will be prohibited. We will ask the participants to report all medications taken during the study at each visit, and record the names, duration, and dosage of the drugs on the case report form (CRF).
Participants will be required to return the unused trial drugs at the next visit. The number of returned drugs will be assessed to evaluate drug compliance, and participants with less than 80% compliance will be excluded.

Outcome

Primary outcomes will include changes in cognitive function, and changes in imaging biomarkers, before and after taking KGT or placebo granules. For assessing cognitive function, the SNSB will be conducted and changes in SNSB results will be analyzed. For measuring the imaging biomarkers, brain MRI scans will be taken to assess the levels of brain metabolites, neurotransmitters, and cerebral blood flow (CBF).
Secondary outcomes will include the safety of KGT granules, and changes in blood-based biomarkers including proteins (amyloid β (Aβ) and tau) and cholesterols, and a comparison of MRI changes between the two groups using age and genotype as covariates. For safety assessments, blood chemistry tests for AST/ALT, glucose, blood urea nitrogen (BUN), Cr, Na, K, Cl, lactate dehydrogenase (LDH), creatine phosphokinase (CPK), and an electrocardiogram (ECG) will be conducted. Blood biomarkers including Aβ protein, tau protein, high-mobility group box (HMGB), small EDRK-rich factor (SERF) 1A, and cholesterol derivatives will also be measured. APOE genotype testing will be performed. Detailed explanations are shown below.

Assessments

K-MMSE and SNSB

The K-MMSE will be performed at the screening visit and after 12 weeks of medication, to screen for MCI and assess the cognitive status of participants. This is a simple screening test for the longitudinal assessment of general cognition [10].
The full version of the SNSB-II will be measured at baseline and after 24 weeks of medication, to evaluate the effect of Kami Guibi-tang on cognitive function. The change in the mean of the SNSB scores, including the SNSB for dementia (SNSB-D) scores, will be compared between the KGT and placebo groups as a primary outcome.
The SNSB is one of the standardized neuropsychological test batteries, widely used in South Korea, for assessing general cognitive abilities. The test is composed of multiple subtests, which evaluate five cognitive aspects: attention, memory, language, visuospatial function, and executive function. Other related tests are included, such as the K-MMSE, Clinical Dementia Rating (CDR), Barthel–Activities of Daily Living (BADL), and Geriatric Depression Scale (GDS). The estimated completion time of the whole battery is 1.5–2 h [11].
A modified version of the original SNSB for dementia (SNSB-D) will be used to provide a general cognitive functioning (GCF) score, drawn from the sum of five domains. The maximum total score is 300 points consisting of 17/300 (6%) for attention, 27/300 (9%) for language and related function, 36/300 (12%) for visuospatial function, 150/300 (50%) for memory, and 70/300 (23%) for frontal/executive function [12]. The contents of the SNSB-D are shown in Table 2.
Table 2
Contents of the Seoul Neuropsychological Battery for Dementia (SNSB-D)
Domains (total score)
Subtests
Maximum points
Attention (17)
Digit span forward
9
Digit span backward
8
Language and related function (27)
K-BNT
15
Calculation
12
Visuospatial function (36)
RCFT copy
36
Memory (150)
Orientation
6
SVLT free/delayed recall
48
SVLT recognition
12
RCFT free/delayed recall
72
RCFT recognition
12
Frontal/executive function (70)
Motor impersistence
3
Contrasting program
3
Go-no-go test
3
Fist-edge-palm
3
Luria loop
3
Categoric word generation
20
Phonemic word generation
15
Stroop test-color reading
20
GCF score
 
300
K-BNT Korean short version of the Boston Naming Test, RCFT Rey Complex Figure Test, SVLT Seoul Verbal Learning Test, GCF global cognitive function

MRI

Participants will undergo brain MRI at baseline and after 24 weeks of medication, to measure changes in brain metabolite and neurotransmitter levels, cerebral blood flow, tissue volume, and to evaluate brain abnormalities.
Proton magnetic resonance spectroscopy (1H-MRS) will be used to measure brain metabolite and neurotransmitter levels. Single-voxel Point-RESolved Spectroscopy (PRESS) MRS will be performed at the precuneus and posterior cingulate area of the brain, with a voxel size of 30 mm × 30 mm × 30 mm, to detect N-acetylaspartate (NAA) and glutamate-glutamine complex (Glx, with both Glu and Gln). N-acetylaspartate and Glx will be quantified using LCModel software. MEshcher-GArwood (MEGA) PRESS MRS will be performed to detect gamma-aminobutyric acid (GABA) in the same area as the PRESS MRS. The amount of GABA will be quantified using GANNET software. Pseudo-continuous arterial spin-labeling (pCASL) MRI will be performed to measure cerebral blood flow (CBF) in the brain. Voxel-based CBF will be mapped using local software. A three-dimensional T1-weighted image will be acquired using the magnetization-prepared rapid gradient-echo (MPRAGE) sequence, to quantify the gray and white matter tissue volume in the brain. T2-weighted turbo-spin echo (TSE), and fluid attenuation inversion recovery (FLAIR) sequences, will be used to evaluate brain abnormalities in participants. Changes in CBF and mean levels of NAA/Cr, Glx/Cr, and GABA/Cr, will be compared between KGT and placebo groups, as a primary outcome.

Blood testing

Amyloid β protein, tau protein, HMGB, SERF 1A, and cholesterol derivatives will be measured in the blood at baseline and after 24 weeks of medication to observe changes in serum proteins and cholesterol associated with AD pathology. Changes in mean values of blood biomarker levels will be compared between the KGT and placebo groups.

Genotyping

Apolipoprotein E (APOE) epsilon genotyping (e23, e33, e34, e44) will be performed using blood samples at baseline, to determine if the effect of KGT is influenced by APOE genotype. The changes in mean values of NAA/Cr, Glx/Cr, GABA/Cr, and CBF before and after the intervention will be compared between carriers and non-carriers of the APOE epsilon 4 allele.

Safety assessment

Laboratory tests and ECGs will be performed for safety outcomes. Blood levels of AST/ALT, glucose, BUN, Cr, Na, K, Cl, cholesterol, LDH, and CPK will be measured at baseline, 12, and 24 weeks after medication. An ECG will be performed at baseline, and 24 weeks after medication. Vital signs will be recorded at every visit. Any abnormal results in laboratory tests, ECG or vital signs, will be closely monitored. The investigators will call participants by phone 4 weeks after completion of medication, to monitor occurrence of any adverse events.

Data management and monitoring

Case report forms (CRFs) will be used for each participant to collect relevant data. To promote data quality and accuracy, one trained investigator will complete the CRFs and a second investigator will independently review all CRFs. All documents will be kept at the study site, and the data will be entered and stored in a password-protected computer. All procedures will comply with confidentiality standards for medical data. All documents and collected data will be kept for 3 years after completion of the study, and will then be destroyed. The data management process will be monitored by an independent agent. Only the investigators and the monitoring agent will have access to the dataset. Auditing will be conducted by the Korean Ministry of Food and Drug Safety.

Statistics

Sample size calculation

This clinical trial is a pilot study to examine the feasibility of a full randomized clinical trial of KGT, and to determine the sample magnitude required for large-scale studies. To our knowledge, no prior study has investigated the effect of KGT on MCI. There are no previous data indicating the sample size needed to yield statistically significant results for determining the effect of KGT by the SNSB scores. For pilot trials, the sample size of 10–20 participants per group was suggested by Kieser and Wassmer. [13]. Browne recommended using at least 30 subjects to estimate parameters [14]. Our target sample size is 30, so 38 individuals will be recruited to allow for a 20% dropout rate.

Data analyses

The investigators, along with an independent professional statistician, will perform the data analyses. All statistical analyses will be performed using SPSS for Windows. The effectiveness test will be performed using the intention-to-treat (ITT) principle and the per-protocol (PP) principle. Missing data will be adjusted with the last-observation-carried-forward (LOCF) imputation method. The safety test will be analyzed using the ITT principle, without adjustment. We will not perform interim analysis.
The student’s t test will be used for parametric variable comparisons between the two groups, and the paired t test will be used for intra-group comparisons. The Mann-Whitney U test will be used for non-parametric variable comparisons between the two groups, and the Wilcoxon signed-rank test will be used for intra-group comparisons.
The paired t test will be used to compare changes in MRI measurements, blood proteins, and cholesterol levels, before and after treatment. Correlation analysis will be used to estimate the relationship between the changes in MRI measures and changes in blood test results. Linear regression analysis will be used to compare changes in MRI measures, between the study group and the placebo group, using age and APOE genotype as covariates. Statistical significance will be set at P < 0.05, and all tests will be two-tailed.

Adverse events

An undesirable, unexpected sign, symptom, or disease that occurs during the trial will be identified as an adverse event (AE), regardless of any causal relationship with the study intervention. Adverse events will be checked for at every visit, from assessment of participants’ subjective reports and by objective examination, including blood tests and ECGs. Monitoring will also be performed by phone each week during the intervention period, and 4 weeks after the end of treatment. All AEs will be recorded in the CRF by the site investigator and assessed for severity and causality. Details of each AE will be recorded, including start and end date, feature, duration, severity, and causal relationship to the study medication.
Serious adverse events (SAEs) are defined as illness requiring hospitalization, events that result in persistent or significant disability or incapacity, events deemed life-threatening, death, a congenital anomaly or birth defect, or other important medical conditions. If SAEs occur, study participation will be discontinued, appropriate measures will be taken immediately, and the Institutional Review Board (IRB) will be notified as soon as possible. All occurrences of AEs will be monitored until they subside or stabilize.

Discussion

Alzheimer’s disease has become one of the most serious health concerns, due to increasing life span and aging populations worldwide. The disease is a chronic, progressive neurodegenerative disorder, characterized by pathological changes including neuritic plaques and neurofibrillary tangles, which accumulate years before clinical symptoms manifest [15]. Early treatment of MCI patients could halt or slow down irreversible progression of neurodegeneration to AD [16]. Nevertheless, there is no proven treatment for MCI to date [17]. In this trial, we will explore the potential of KGT as a therapeutic agent for MCI patients.
Clinical symptoms are not evident in MCI, so we will utilize an extensive assessment to evaluate the effect of KGT on cognitive function and disease progression. Instruments, such as the K-MMSE and the Alzheimer’s Disease Assessment Scale-cognitive subscale (ADAS-cog), have been mainly used in previous studies for measuring cognitive decline. However, they lack sensitivity in detecting mild degrees of cognitive dysfunction, which demonstrate ceiling effects in MCI [18]. We will use the full neuropsychological battery, the SNSB, which contains comprehensive and diverse tests with varying difficulties, to sensitively and accurately monitor changes in cognitive function [11].
We will also measure neuroimaging markers that reflect dynamic neuropathological processes. Structural MRI-based measurements of brain atrophy are the most widely accepted marker of AD progression [19]. However, the utility of structural MRI for MCI is limited because overt loss of neurons and associated brain atrophy occur in the later stages of AD [20]. Functional changes also occur in the brains of MCI and AD patients. There is consistent reduction of cerebral metabolism, blood flow, and disturbances in neurotransmission that precede substantial brain atrophy [21]. Therefore, changes in brain metabolite and neurotransmitter levels, and cerebral blood flow will be measured in this trial.
Proton MRS is a sensitive method for estimating brain metabolite and neurotransmitter concentrations [22], including NAA, myo-Inositol (mI), choline (Cho), Glx, and GABA, frequently measured as ratios to creatine [23]. A decrease in concentrations of NAA, a neuronal metabolite, and Glx, a component of the excitatory neurotransmitter system in the brain, is indicative of neuronal dysfunction in AD [24]. Detecting changes in neuronal markers may be suitable for assessing treatment responses.
Arterial spin-labeling perfusion MRI (ASL MRI) is used to quantify regional cerebral blood flow (rCBF) [25]. Alzheimer’s disease patients show regional hypoperfusion that reflects patterns of reduced brain functional activity, using this technique [26]. Studies have suggested that ASL MRI is appropriate for examining neural response to pharmacological agents [27]. Prior studies that measured rCBF used Single Photon Emission Computed Tomography (SPECT). However, ASL MRI is advantageous because there is no exposure to ionizing radiation or the need for intravenously administered contrast agents, and there is higher accessibility, lower cost, and higher spatial resolution [26].
As secondary outcomes, we will observe changes in blood-based biomarkers levels, including Aβ protein, tau protein, HMGB, SERF 1A, and cholesterol derivatives, which are proposed to be associated with AD pathology [28]. Aβ protein and tau proteins are the main feature of AD neuropathology, and are closely associated with neuronal loss. High plasma Aβ levels are a risk factor for AD [29]. High total cholesterol levels accelerate the production of Aβ protein in AD, and are related to increased cognitive impairment in humans [30]. The strongest genetic risk factor for AD, APOE genotype, will also be determined by plasma APOE proteins [31]. This genetic marker will provide information for distinguishing which patients are most likely to benefit from KGT administration.
This pilot study will be the first rigorous clinical analysis of KGT for the treatment of aMCI, and should provide evidence for the effectiveness and safety and of KGT. Our extensive assessment, using various biomarkers, should reveal the mechanisms underlying the effect of KGT on disease progression. The findings will support a large-scale confirmatory clinical trial to gather evidence for KGT use in MCI patients.
There are some limitations in this protocol. The target sample size is relatively small, and the sample size could not be calculated by standard methods. However, this is a pilot study to generate data for sample size calculations in a future randomized trial, which will include a larger sample size. Also, the total intervention period is 24 weeks and the monitoring period after the completion of taking medicine is 4 weeks, which is relatively short considering the nature of the disease. Long-term follow-up will be necessary in future trials.

Trial status

This study began recruitment in May 2017 and we have currently enrolled 33 participants.

Acknowledgements

This study was supported by the Convergence of Conventional Medicine and Traditional Korean Medicine R&D program.
The trial will be carried out in accordance with the Declaration of Helsinki and the Korean Good Clinical Practice Guidelines. The trial has been approved by the Institutional Review Board of Kyung Hee University Hospital at Gangdong (KHNMC-OH-IRB 2016–12–006-006) and the Korean Ministry of Food and Drug Safety (31234), and is registered at the Korean Clinical Trial Registry (Registration number: KCT0002407, URL: http://​cris.​nih.​go.​kr/​). The current protocol version (1.5) was approved on 13 December 2018, and developed according to the Standard Protocol Items: Recommendations for Interventional Trials (SPIRIT) Statement (see Additional file 1). All items from the World Health Organization Trial Registration Data Set have been drawn. Any modifications to the protocol will be approved by the Institutional Review Board before being implemented. Signed informed consent will be obtained from all qualified participants. Participation in this study is completely voluntary, and participants can withdraw from the study any time.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Petersen RC, Caracciolo B, Brayne C, Gauthier S, Jelic V, Fratiglioni L. Mild cognitive impairment: a concept in evolution. J Intern Med. 2014;275(3):214–28.CrossRef Petersen RC, Caracciolo B, Brayne C, Gauthier S, Jelic V, Fratiglioni L. Mild cognitive impairment: a concept in evolution. J Intern Med. 2014;275(3):214–28.CrossRef
2.
Zurück zum Zitat Petersen RC, Doody R, Kurz A, Mohs RC, Morris JC, Rabins PV, et al. Current concepts in mild cognitive impairment. Arch Neurol. 2001;58:1985–92.CrossRef Petersen RC, Doody R, Kurz A, Mohs RC, Morris JC, Rabins PV, et al. Current concepts in mild cognitive impairment. Arch Neurol. 2001;58:1985–92.CrossRef
3.
Zurück zum Zitat Petersen RC, Thomas RG, Grundman M, et al. Vitamin E and donepezil for the treatment of mild cognitive impairment. N Engl J Med. 2005;352(23):2379–88.CrossRef Petersen RC, Thomas RG, Grundman M, et al. Vitamin E and donepezil for the treatment of mild cognitive impairment. N Engl J Med. 2005;352(23):2379–88.CrossRef
4.
Zurück zum Zitat Winblad B, Wimo A, Engedal K, Soininen H, Verhey F, Waldemar G, et al. 3-year study of donepezil therapy in Alzheimer’s disease: effects of early and continuous therapy. Dement Geriatr Cogn Disord. 2006;21(5–6):353–63.CrossRef Winblad B, Wimo A, Engedal K, Soininen H, Verhey F, Waldemar G, et al. 3-year study of donepezil therapy in Alzheimer’s disease: effects of early and continuous therapy. Dement Geriatr Cogn Disord. 2006;21(5–6):353–63.CrossRef
5.
Zurück zum Zitat Saunders NL, Summers MJ. Attention and working memory deficits in mild cognitive impairment. J Clin Exp Neuropsychol. 2010;32(4):350–7.CrossRef Saunders NL, Summers MJ. Attention and working memory deficits in mild cognitive impairment. J Clin Exp Neuropsychol. 2010;32(4):350–7.CrossRef
6.
Zurück zum Zitat Nishizawa K, Saito H, Nishiyama N. Effects of Kamikihi-To, a traditional Chinese medicine, on passive and conditioned avoidance performance impairment in senescence accelerated mouse (SAM). Jpn J Pharmacol. 1990;54:375–82.CrossRef Nishizawa K, Saito H, Nishiyama N. Effects of Kamikihi-To, a traditional Chinese medicine, on passive and conditioned avoidance performance impairment in senescence accelerated mouse (SAM). Jpn J Pharmacol. 1990;54:375–82.CrossRef
7.
Zurück zum Zitat Egashira N, Manome N, Kurauchi K, et al. Kamikihi-to, a Kampo medicine, ameliorates impairment of spatial memory in rats. Phytother Res. 2007;21:126–9.CrossRef Egashira N, Manome N, Kurauchi K, et al. Kamikihi-to, a Kampo medicine, ameliorates impairment of spatial memory in rats. Phytother Res. 2007;21:126–9.CrossRef
8.
Zurück zum Zitat Tohda C, Nakada R, Urano T, Okonogi A, Kuboyama T. Kamikihi-to (KKT) rescues axonal and synaptic degeneration associated with memory impairment in a mouse model of Alzheimer’s disease, 5XFAD. Int J Neurosci. 2011;121:641–8.CrossRef Tohda C, Nakada R, Urano T, Okonogi A, Kuboyama T. Kamikihi-to (KKT) rescues axonal and synaptic degeneration associated with memory impairment in a mouse model of Alzheimer’s disease, 5XFAD. Int J Neurosci. 2011;121:641–8.CrossRef
9.
Zurück zum Zitat Higashi K, Rakugi H, Yu H, Moriguchi A, Shintani T, Ogihara T. Effect of kihito extract granules on cognitive function in patients with Alzheimer’s-type dementia. Geriatr Gerontol Int. 2007;7:245–51.CrossRef Higashi K, Rakugi H, Yu H, Moriguchi A, Shintani T, Ogihara T. Effect of kihito extract granules on cognitive function in patients with Alzheimer’s-type dementia. Geriatr Gerontol Int. 2007;7:245–51.CrossRef
10.
Zurück zum Zitat Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”: a practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12:189–98.CrossRef Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”: a practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12:189–98.CrossRef
11.
Zurück zum Zitat Kang Y, Na DL. Seoul Neuropsychological Screening Battery (SNSB). Seoul: Human Brain Research & Consulting Co; 2003. Kang Y, Na DL. Seoul Neuropsychological Screening Battery (SNSB). Seoul: Human Brain Research & Consulting Co; 2003.
12.
Zurück zum Zitat Ahn H-J, Chin J, Park A, Lee BH, et al. Seoul Neuropsychological Screening Battery-Dementia Version(SNSB-D): a useful tool for assessing and monitoring cognitive impairments in dementia patients. J Korean Med Sci. 2010;25:1071–6.CrossRef Ahn H-J, Chin J, Park A, Lee BH, et al. Seoul Neuropsychological Screening Battery-Dementia Version(SNSB-D): a useful tool for assessing and monitoring cognitive impairments in dementia patients. J Korean Med Sci. 2010;25:1071–6.CrossRef
13.
Zurück zum Zitat Kieser M, Wassmer G. On the use of the upper confidence limit for the variance from a pilot sample for sample size determination. Biom J. 1996;38:941–9.CrossRef Kieser M, Wassmer G. On the use of the upper confidence limit for the variance from a pilot sample for sample size determination. Biom J. 1996;38:941–9.CrossRef
14.
Zurück zum Zitat Browne RH. On the use of a pilot sample for sample size determination. Stat Med. 1995;14:1933–40.CrossRef Browne RH. On the use of a pilot sample for sample size determination. Stat Med. 1995;14:1933–40.CrossRef
15.
Zurück zum Zitat Heininger K. A unifying hypothesis of Alzheimer’s disease II. Pathophysiological processes. Hum Psychopharmacol Clin Exp. 1999;14:525–81.CrossRef Heininger K. A unifying hypothesis of Alzheimer’s disease II. Pathophysiological processes. Hum Psychopharmacol Clin Exp. 1999;14:525–81.CrossRef
16.
Zurück zum Zitat Gauthier S, Reisberg B, Zaudig M, et al. Mild cognitive impairment. Lancet. 2006;367:1262–70.CrossRef Gauthier S, Reisberg B, Zaudig M, et al. Mild cognitive impairment. Lancet. 2006;367:1262–70.CrossRef
17.
Zurück zum Zitat Tricco AC, Soobiah C, Berliner S, et al. Efficacy and safety of cognitive enhancers for patients with mild cognitive impairment: a systematic review and meta-analysis. CMAJ. 2013;185:1393.CrossRef Tricco AC, Soobiah C, Berliner S, et al. Efficacy and safety of cognitive enhancers for patients with mild cognitive impairment: a systematic review and meta-analysis. CMAJ. 2013;185:1393.CrossRef
18.
Zurück zum Zitat Kim J, Na HK, Byun J, et al. Tracking cognitive decline in amnestic mild cognitive impairment and early-stage Alzheimer dementia: Mini-Mental State Examination versus Neuropsychological Battery. Dement Geriatr Cogn Disord. 2017;44:1–2.CrossRef Kim J, Na HK, Byun J, et al. Tracking cognitive decline in amnestic mild cognitive impairment and early-stage Alzheimer dementia: Mini-Mental State Examination versus Neuropsychological Battery. Dement Geriatr Cogn Disord. 2017;44:1–2.CrossRef
19.
Zurück zum Zitat Kantarci K, Jack C Jr. Quantitative magnetic resonance techniques as surrogate markers in Alzheimer’s disease. NeuroRx J Am Soc Exp Neurotherap. 2004;1:196–205. Kantarci K, Jack C Jr. Quantitative magnetic resonance techniques as surrogate markers in Alzheimer’s disease. NeuroRx J Am Soc Exp Neurotherap. 2004;1:196–205.
20.
Zurück zum Zitat Citron M. Alzheimer’s disease: strategies for disease modification. Nat Rev Drug Discov. 2010;9:387–8.CrossRef Citron M. Alzheimer’s disease: strategies for disease modification. Nat Rev Drug Discov. 2010;9:387–8.CrossRef
21.
Zurück zum Zitat Hata S, Suzuki T, et al. Utility of SPM8 plus DARTEL (VSRAD) combined with magnetic resonance spectroscopy as adjunct techniques for screening and predicting dementia due to Alzheimer’s disease in clinical practice. J Alzheimers Dis. 2014;41:1207–22.CrossRef Hata S, Suzuki T, et al. Utility of SPM8 plus DARTEL (VSRAD) combined with magnetic resonance spectroscopy as adjunct techniques for screening and predicting dementia due to Alzheimer’s disease in clinical practice. J Alzheimers Dis. 2014;41:1207–22.CrossRef
22.
Zurück zum Zitat Jessen F, Gür O, Block W, Ende G, Frölich L, Hammen T, et al. A multicenter (1)H-MRS study of the medial temporal lobe in AD and MCI. Neurology. 2009;72:1735–40.CrossRef Jessen F, Gür O, Block W, Ende G, Frölich L, Hammen T, et al. A multicenter (1)H-MRS study of the medial temporal lobe in AD and MCI. Neurology. 2009;72:1735–40.CrossRef
23.
Zurück zum Zitat Londono AC, Castellanos FX, Arbelaez A, et al. An 1H-MRS framework predicts the onset of Alzheimer’s disease symptoms in PSEN1 mutation carriers. Alzheimers Dement. 2014;10:552–61.CrossRef Londono AC, Castellanos FX, Arbelaez A, et al. An 1H-MRS framework predicts the onset of Alzheimer’s disease symptoms in PSEN1 mutation carriers. Alzheimers Dement. 2014;10:552–61.CrossRef
24.
Zurück zum Zitat Block W, Träber F, Flacke S, et al. In-vivo proton MR-spectroscopy of the human brain: assessment of N-acetylaspartate (NAA) reduction as a marker for neurodegeneration. Amino Acids. 2002;23(1):317–23.CrossRef Block W, Träber F, Flacke S, et al. In-vivo proton MR-spectroscopy of the human brain: assessment of N-acetylaspartate (NAA) reduction as a marker for neurodegeneration. Amino Acids. 2002;23(1):317–23.CrossRef
25.
Zurück zum Zitat Alsop DC, Dai W, Grossman M, Detre JA. Arterial spin labeling blood flow MRI: its role in the early characterization of Alzheimer’s disease. J Alzheimers Dis. 2010;20:871.CrossRef Alsop DC, Dai W, Grossman M, Detre JA. Arterial spin labeling blood flow MRI: its role in the early characterization of Alzheimer’s disease. J Alzheimers Dis. 2010;20:871.CrossRef
26.
Zurück zum Zitat Wolk DA, Detre JA. Arterial spin labeling MRI: an emerging biomarker for Alzheimer’s disease and other neurodegenerative conditions. Curr Opin Neurol. 2012;25:421.CrossRef Wolk DA, Detre JA. Arterial spin labeling MRI: an emerging biomarker for Alzheimer’s disease and other neurodegenerative conditions. Curr Opin Neurol. 2012;25:421.CrossRef
27.
Zurück zum Zitat Alsop DC, Detre JA, Grossman M. Assessment of cerebral blood flow in Alzheimer’s disease by spin-labeled magnetic resonance imaging. Ann Neurol. 2000;47:93–100.CrossRef Alsop DC, Detre JA, Grossman M. Assessment of cerebral blood flow in Alzheimer’s disease by spin-labeled magnetic resonance imaging. Ann Neurol. 2000;47:93–100.CrossRef
28.
Zurück zum Zitat Schneider P, Hampel H, Buerger K. Biological marker candidates of Alzheimer’s disease in blood, plasma, and serum. CNS Neurosci Ther. 2009;15:358–74.CrossRef Schneider P, Hampel H, Buerger K. Biological marker candidates of Alzheimer’s disease in blood, plasma, and serum. CNS Neurosci Ther. 2009;15:358–74.CrossRef
29.
Zurück zum Zitat Schupf N, Tang MX, Fukuyama H, et al. Peripheral Abeta subspecies as risk biomarkers of Alzheimer’s disease. Proc Natl Acad Sci. U S A. 2008;105:14052–7.CrossRef Schupf N, Tang MX, Fukuyama H, et al. Peripheral Abeta subspecies as risk biomarkers of Alzheimer’s disease. Proc Natl Acad Sci. U S A. 2008;105:14052–7.CrossRef
30.
Zurück zum Zitat Yaffe K, Barrett-Connor E, Lin F, Grady D. Serum lipoprotein levels, statin use, and cognitive function in older women. Arch Neurol. 2002;59:378–84.CrossRef Yaffe K, Barrett-Connor E, Lin F, Grady D. Serum lipoprotein levels, statin use, and cognitive function in older women. Arch Neurol. 2002;59:378–84.CrossRef
31.
Zurück zum Zitat Hsiung GY, Sadovnick AD, Feldman H. Apolipoprotein E epsilon4 genotype as a risk factor for cognitive decline and dementia: data from the Canadian Study of Health and Aging. CMAJ. 2004;171:863–7.CrossRef Hsiung GY, Sadovnick AD, Feldman H. Apolipoprotein E epsilon4 genotype as a risk factor for cognitive decline and dementia: data from the Canadian Study of Health and Aging. CMAJ. 2004;171:863–7.CrossRef
Metadaten
Titel
The effectiveness and safety of Kami Guibi-tang for mild cognitive impairment: study protocol of a pilot, randomized, placebo-controlled, double-blind trial
verfasst von
Hee-Yeon Shin
Jeong-Hwa Kim
Geon-Ho Jahng
Woo-Sang Jung
Seong-Uk Park
Chang-Nam Ko
Jung-Mi Park
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Trials / Ausgabe 1/2019
Elektronische ISSN: 1745-6215
DOI
https://doi.org/10.1186/s13063-019-3567-1

Weitere Artikel der Ausgabe 1/2019

Trials 1/2019 Zur Ausgabe