WO2015147742A1 - Use of benzoic acid salt in the manufactue of a composition for preventing or treating dementia or mild cognitive impairment - Google Patents

Use of benzoic acid salt in the manufactue of a composition for preventing or treating dementia or mild cognitive impairment Download PDF

Info

Publication number
WO2015147742A1
WO2015147742A1 PCT/SG2014/000140 SG2014000140W WO2015147742A1 WO 2015147742 A1 WO2015147742 A1 WO 2015147742A1 SG 2014000140 W SG2014000140 W SG 2014000140W WO 2015147742 A1 WO2015147742 A1 WO 2015147742A1
Authority
WO
WIPO (PCT)
Prior art keywords
dementia
day
alzheimer
cognitive impairment
week
Prior art date
Application number
PCT/SG2014/000140
Other languages
French (fr)
Inventor
Hsien-Yuan Lane
Chieh-Hsin Lin
Original Assignee
Kaohsiung Chang Gung Memorial Hospital
China Medical University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Kaohsiung Chang Gung Memorial Hospital, China Medical University filed Critical Kaohsiung Chang Gung Memorial Hospital
Priority to SG11201507188QA priority Critical patent/SG11201507188QA/en
Priority to KR1020177024853A priority patent/KR102162073B1/en
Priority to KR1020167003184A priority patent/KR20160029837A/en
Priority to PCT/SG2014/000140 priority patent/WO2015147742A1/en
Priority to DE212014000063.7U priority patent/DE212014000063U1/en
Priority to AU2014386718A priority patent/AU2014386718B8/en
Priority to JP2016510654A priority patent/JP2016517866A/en
Priority to CA2902498A priority patent/CA2902498C/en
Publication of WO2015147742A1 publication Critical patent/WO2015147742A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/166Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the carbon of a carboxamide group directly attached to the aromatic ring, e.g. procainamide, procarbazine, metoclopramide, labetalol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/216Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acids having aromatic rings, e.g. benactizyne, clofibrate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Definitions

  • the present invention relates generally to a novelty treatment for dementia or mild cognitive impairment, and more specifically to a use of benzoic acid salt in the manufacture of a composition for preventing or treating dementia or mild cognitive impairment.
  • AD Alzheimer's disease
  • MCI Mild cognitive impairment
  • aMCI amnestic MCI
  • AChEI acetylcholine esterase inhibitor
  • NMDAR activity is essential for cognitive function, its role in AD is still not fully understood. NMDAR over-activation by glutamate results in cell death. The excitotoxicity is one of the theories of AD, particularly in the late stage (54). Based on the hypothesis of NMDAR overactivation (7), NMDAR antagonists are developed for the treatment of AD. Memantine is an uncompetitive NMDAR partial antagonist of low affinity, which supposedly can block NMDAR overactivation by preventing excessive influx of calcium (8-10) and has been used for the treatment of moderate-severe AD. However, it has limited efficacy at the early phase, including MCI and mild AD (12). NMDAR antagonists such as MK-801 also induce apoptosis and neurodegeneration in both in vitro and in vivo studies (13). Ketamine, another NMDAR antagonist, impaired spatial learning and verbal
  • Optimal NMDAR activation is pivotal for synaptic plasticity (15), memory and cognitive function (16). Attenuation of NMDAR-mediated neurotransmission can result in loss of neuronal plasticity and cognitive deficits in the aging brain, which may account for clinical deterioration and brain atrophy (17). Age-related decrease in the density of NMDAR in cerebral cortex and hippocampus was observed in humans (18). Earlier studies also found a decrease of glycine-dependent radioligand binding to the NMDAR in cerebral cortices from post-mortem and neurosurgical tissues in patients with AD (19, 20).
  • D-cycloserine a partial agonist at the glycine site of NMDAR, was reported in some clinical studies to activate the NMDAR in brains of AD patients (21) and improve their score on the cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-cog) (22).
  • NMDAR enhancement be beneficial for early and mild dementia.
  • Lower levels of D-serine and higher levels of L-serine in the serum were also observed in patients with AD (56). Therefore, in addition to the cholinergic system, dysfunction of NMD A neurotransmission may also play an important role in the pathophysiology of AD.
  • D-amino acids oxidase DAAO
  • D-amino acids oxidase 24-26
  • D-amino acids oxidase 24-26
  • D-amino acids oxidase 24-26
  • D-amino acids oxidase 24-26
  • D-amino acids oxidase 24-26
  • D-amino acids oxidase 24-26
  • D-amino acids oxidase a flavoenzyme of peroxisomes responsible for degrading D-serine and D-alanine
  • D-amino acids which are the neurotransmitters for the coagonist site of the NMDAR.
  • Recent data indicate that aging is related with reduced D-serine levels and thereby impaired NMDAR transmission, and D-serine treatment significantly decreases the extent of neuron death, suggesting that D-serine has neuroprotective effect against apoptosis (27).
  • Enhancing NMDAR through DAAO inhibition may be a safe way to reduce nephrotoxicity of D-serine (29), particularly in the elderly population.
  • Sodium benzoate is a DAAO inhibitor.
  • Benzoic acid exists in many plants and is a natural constituent of food, including milk products (30).
  • Benzoic acid and its salts including sodium benzoate, which are generally recognized as safe (GRAS) are also food preservatives widely used in manufacturing fruit jelly, buffer, soy-bean sauce, processed meat, etc. (31).
  • NMDAR N-methyl-D-aspartate receptor
  • NMDA enhancing agents may be beneficial for the early declining process of AD and mild ⁇ o ⁇ kivejmpairmeiit
  • the present invention provides a use of benzoic acid salt in the manufacture of a composition for preventing or treating dementia or mild cognitive impairment.
  • the benzoic acid salt can be sodium benzoate, potassium benzoate or calcium benzoate, and preferably, the benzoic acid salt is sodium benzoate.
  • an effective amount of benzoic acid salt can be 200 milligrams (mg)/day to 2000 mg/day, preferably 500 mg/day to 900 mg/day, and more preferably 750 mg/day.
  • an effective amount of sodium benzoate is 200 mg/day to 2000 mg/day, preferably 500 mg/day to 900 mg/day, and more preferably 750 mg/day.
  • the dementia includes early-phase dementia. In one embodiment of the present application, the early-phase dementia includes mild Alzheimer's disease.
  • the mild cognitive impairment includes amnestic mild cognitive impairment.
  • FIG. 1 shows a flow diagram and disposition for two treatment groups.
  • the term “dementia” refers to a group of symptoms affecting intellectual and social abilities severe enough to interfere with daily functioning, including memory loss, language problems, inability to learn or remember new information, etc (78).
  • the term “early-phase dementia” refers to the condition of dementia patients whose CDR (Clinical Dementia Rating) score is not more than 1.
  • AD Alzheimer's disease
  • CDR Chronic Dementia Rating
  • MCI cognitive impairment
  • CDR Clinical Dementia Rating
  • aMCI amnestic mild cognitive impairment
  • CDR Clinical Dementia Rating
  • DAAO sodium benzoateT ⁇ D ⁇ ino acid oxidase
  • NINCDS-ADRDA National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer's Disease and Related Disorders Association
  • CDR Clinical Dementia Rating
  • MCI Mini-Mental State Examination
  • AChEI For patients who had already been on AChEI therapy, AChEI had to be continued for at least three months before enrollment. AChEI dose had to be kept unchanged during the study duration. For patients who had not yet been on AChEI therapy, AChEI or other anti-dementia medication was forbidden during the study duration.
  • Exclusion criteria included history of significant cerebrovascular disease; Hachinski Ischemic Score >4; major neurological, psychiatric or medical conditions other than AD; substance (including alcohol) abuse or dependence; delusion, hallucination or delirium symptoms; severe visual or hearing loss; and inability to follow protocol.
  • ADAS-cog Alzheimer's disease assessment scale-cognitive subscale (43) measured at weeks 0, 8, 16, and 24.
  • ADAS-cog is the most popular cognitive assessment instrument used in At) clinical trials. It consists of 11 tasks, including word recall, naming, commands, constructional praxis, ideational praxis, orientation, word recognition, instructions remembering, spoken language ability, word-finding difficulty and comprehension. Its scores range from 0 (best) to 70 (worst).
  • the secondary outcome measurements included the Clinician's Interview-Based Impression of Change plus Caregiver Input (CIBIC-plus) (44) measured at weeks 8, 16 and 24, and the additional cognition composite measured at baseline and endpoint (was measured at the end of each patent's additional cognition composite measurement).
  • CBIC-plus Clinician's Interview-Based Impression of Change plus Caregiver Input
  • CIBIC-plus is a global assessment of change based on a comprehensive, semi-structured interview which includes caregiver-supplied information. It is a 7-point rating scale ranging from 1-7, where 1 represents markedly improved; 4, no change; and 7, markedly worse.
  • the additional cognition composite was calculated by the average of the T scores of speed of processing (Category Fluency), working memory (WMS-III, Spatial Span) (45) and verbal learning and memory tests (WMS-III, Word Listing) (45).
  • the raw score of speed of processing, working memory and verbal learning and memory tests was standardized to a T score with a mean of 50 and a standard deviation of 10 for making each test comparative.
  • the additional cognition composite was applied in combination with ADAS-cog to make the cognitive assessment more thorough. Decrease in processing speed has been found to be associated with aging (46, 47).
  • Working memory (48) and verbal learning/memory (49) also decline in patients with AD.
  • Rivastigmine (dose, mean ⁇ SD) 0 (0.0 ⁇ 0.0) 3 (7.5 ⁇ 2.6) NA
  • AChEI acetylcholine esterase inhibitor
  • BMI body mass index
  • CDR Clinical Dementia Rating
  • NA not associated. a Fisher's exact test.
  • Additional cognition composite the composite test score of speed of processing, working memory, and verbal learning, and memory.
  • the results were similar when the baseline ADAS-cog score was controlled in the GEE model (as shown in Table SI).
  • ADAS-cog Alzheimer's disease assessment scale-cognitive subscale.
  • ADAS-cog is widely used in AD clinical trials, it may be less sensitive for MCI (67).
  • One of the strategies to improve the detection of responsiveness for MCI is to add additional cognitive tests. People with MCI have been found to be impaired in neuropsychological functions (68) such as speed of processing (69), working memory (70) and verbal learning and memory (71).
  • neuropsychological functions 68
  • 69 speed of processing
  • 70 working memory
  • 70 verbal learning and memory
  • sodium benzoate showed borderline significance in improving the additional cognition composite, consisting of speed of processing, working memory, and verbal learning/memory, but not in ADAS-cog score.
  • Our result echoes the suggestion that additional neuropsychological tests which are more sensitive to subtle deficits should also be applied in the trials for MCI.
  • Endpoint x drug 10.0 ⁇ 6.6 (15) 15.4 + 11.3 (14) -5.4755 2.1031 -2.60 0.0092
  • Estimatimate is the coefficient of treatment-visit interaction term in the GEE method multiple linear regression model. A first-order autoregressive covariance matrix was fit to the within-patient repeated measures. The p values were based on two-tailed tests.
  • CD Clinical Dementia Rating
  • the present invention is the first to apply a DAAO inhibitor, sodium benzoate herein, as a novel treatment for the early stage of cognitive decline.
  • a DAAO inhibitor sodium benzoate herein
  • the result showed that sodium benzoate had better efficacy than placebo in improving ADAS-cog score, additional cognition composite (consisting of speed of processing, working memory, verbal learning and memory) and global function in all subjects as a whole.
  • sodium benzoate showed borderline significance in improving the cognition composite.
  • sodium benzoate also demonstrated favorable safety profiles.
  • sodium benzoate provided better efficacy than placebo at week 16 and week 24, with the mean dose of 525 mg/day and 716 mg/day respectively, possibly implying that sodium benzoate at 500-750 mg/day is more effective than 250 mg/day. Another possibility is that longer sodium benzoate treatment duration yields better treatment response.
  • AChEIs are commonly used for the treatment of AD (57, 58), but are not recommended for the treatment of MCI due to weak beneficial effects and risk of side effects (59, 60).
  • the consensus statement from the British Association for Psychopharmacology concludes that neither AChEIs nor memantine is effective in treating MCI (61).
  • Other compounds commonly used for the treatment of MCI such as vitamin E (62), folic acid (63), omega-3 fatty acid (64), piracetam (65) and Ginkgo biloba (66), also failed to show convincing evidence for a cognitive enhancing effect.
  • Sodium benzoate is generally safe and its efficacy for aMCI reached a trend of improvement in the current small-sized study.
  • the present invention suggests that sodium benzoate, a DAAO inhibitor, is beneficial for cognitive and overall function in patients with early-phase AD and potentially beneficial for aMCI.
  • the use of sodium benzoate for early AD and aMCI will bring hope for the growing aging population with cognitive decline.
  • benzoate treatment for early-phase dementia may be due to activation of neurogenesis and anti-apoptosis.
  • Procter AW Stirling JM, Stratmann GC, Cross AJ, Bowen DM (1989): Loss of glycine-dependent radioligand binding to the N-methyl-D-aspartate-phencyclidine receptor complex in patients with Alzheimer's disease. Neurosci Lett. 101 :62-66.
  • Vanoni MA Cosma A, Mazzeo D, Mattevi A, Todone F, Curti B (1997): Limited proteolysis and X-ray crystallography reveal the origin of substrate specificity and of the rate-limiting product release during oxidation of D-amino acids catalyzed by mammalian D-amino acid oxidase. Biochemistry. 36:5624-5632.
  • Oxidase (DAAO) Inhibitors The open medicinal chemistry journal. 4:3-9.
  • Salthouse TA (1996): The processing-speed theory of adult age differences in cognition. Psychological review. 103:403-428.
  • Neuropsychiatrie Deutschen, Diagnostik, Therapie und Rehabilitation : Organ der Gesellschafi Oeckischer Nervenazie und Psychiater. 21 :230-233.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Neurology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Emergency Medicine (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The present invention provides a novel use of benzoic acid salt in the manufacture of a composition for preventing or treating dementia or mild cognitive impairment, particularly, for preventing or treating mild Alzheimer's disease or amnestic mild cognitive impairment.

Description

USE OF BENZOIC ACID SALT IN THE MANUFACTUE OF A COMPOSITION FOR PREVENTING OR TREATING DEMENTIA OR MILD COGNITIVE IMPAIRMENT
BACKGROUND OF THE INVENTION FIELD OF THE INVENTION
The present invention relates generally to a novelty treatment for dementia or mild cognitive impairment, and more specifically to a use of benzoic acid salt in the manufacture of a composition for preventing or treating dementia or mild cognitive impairment.
Description of Related Art The prevalence of dementia in the elderly is increasing rapidly in the aging society, of which the deteriorating clinical course is a heavy burden to both the patients and their family. Early detection and intervention of Alzheimer's disease (referred to as "AD" hereinafter) is pivotal for the outcome (1). Mild cognitive impairment (referred to as "MCI" hereinafter), particularly amnestic MCI (referred to as "aMCI" hereinafter), is a risk factor and may be a prodromal stage of AD. The mainstream treatment for mild and moderate AD is acetylcholine esterase inhibitor (referred to as "AChEI" hereinafter). However, its efficacy and tolerability are unsatisfactory. Besides, AChEI does not show convincing efficacy for MCI (2-4), implying that other mechanism(s) may underlie the pathogenesis of MCI.
Although NMDAR activity is essential for cognitive function, its role in AD is still not fully understood. NMDAR over-activation by glutamate results in cell death. The excitotoxicity is one of the theories of AD, particularly in the late stage (54). Based on the hypothesis of NMDAR overactivation (7), NMDAR antagonists are developed for the treatment of AD. Memantine is an uncompetitive NMDAR partial antagonist of low affinity, which supposedly can block NMDAR overactivation by preventing excessive influx of calcium (8-10) and has been used for the treatment of moderate-severe AD. However, it has limited efficacy at the early phase, including MCI and mild AD (12). NMDAR antagonists such as MK-801 also induce apoptosis and neurodegeneration in both in vitro and in vivo studies (13). Ketamine, another NMDAR antagonist, impaired spatial learning and verbal
Figure imgf000004_0001
(14). These findings raise concern that NMD A antagonist may impair cognition and memory in early AD.
Optimal NMDAR activation is pivotal for synaptic plasticity (15), memory and cognitive function (16). Attenuation of NMDAR-mediated neurotransmission can result in loss of neuronal plasticity and cognitive deficits in the aging brain, which may account for clinical deterioration and brain atrophy (17). Age-related decrease in the density of NMDAR in cerebral cortex and hippocampus was observed in humans (18). Earlier studies also found a decrease of glycine-dependent radioligand binding to the NMDAR in cerebral cortices from post-mortem and neurosurgical tissues in patients with AD (19, 20). D-cycloserine, a partial agonist at the glycine site of NMDAR, was reported in some clinical studies to activate the NMDAR in brains of AD patients (21) and improve their score on the cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-cog) (22).
The current study suggests that NMDAR enhancement be beneficial for early and mild dementia. There is an age-related decrease of glutamate content and synthesis in human cerebral cortex and hippocampus (18, 55), of which the most significant and consistent finding is decreased density of NMDAR in the elderly and in patients with AD (18). Lower levels of D-serine and higher levels of L-serine in the serum were also observed in patients with AD (56). Therefore, in addition to the cholinergic system, dysfunction of NMD A neurotransmission may also play an important role in the pathophysiology of AD.
There are several avenues to enhance NMDA activation. One of them is inhibiting the activity of D-amino acids oxidase (DAAO), a flavoenzyme of peroxisomes responsible for degrading D-serine and D-alanine (24-26), and thereby raising levels of the D-amino acids which are the neurotransmitters for the coagonist site of the NMDAR. Recent data indicate that aging is related with reduced D-serine levels and thereby impaired NMDAR transmission, and D-serine treatment significantly decreases the extent of neuron death, suggesting that D-serine has neuroprotective effect against apoptosis (27). In addition, neural stem cells from postnatal mouse forebrain can synthesize D-serine and, thereby, stimulate proliferation and neuronal differentiation of the stem cells (28).
Enhancing NMDAR through DAAO inhibition may be a safe way to reduce nephrotoxicity of D-serine (29), particularly in the elderly population. Sodium benzoate is a DAAO inhibitor. Benzoic acid exists in many plants and is a natural constituent of food, including milk products (30). Benzoic acid and its salts including sodium benzoate, which are generally recognized as safe (GRAS), are also food preservatives widely used in manufacturing fruit jelly, buffer, soy-bean sauce, processed meat, etc. (31).
There are several other preclinical studies supporting the CNS effects of DAAO inhibitors, though the memory effect was not examined (32-34). N-methyl-D-aspartate receptor (NMDAR)-mediated neurotransmission is vital for learning and memory. Hypofunction of NMDAR has been reported to play a role in the pathophysiology of Alzheimer's disease (AD), particularly in the early phase. Enhancing NMDAR activation may be a novel treatment approach. One of the methods to enhance NMDAR activity is to raise the levels of NMDA coagonists by blocking their metabolism. Sodium benzoate is effective in NMDAR models such as pain relief (35,36) and partially prevented cell death in glial cells (37). CNS bioavailability of benzoate is good (38). To test whether that DAAO inhibition is beneficial for the early phase of dementia, the inventors conducted this trial to examine the efficacy and safety of sodium benzoate in patients with aMCI or mild AD. SUMMARY OF THE INVENTION
On account of the supporting evidence, the inventor proposed that NMDA enhancing agents may be beneficial for the early declining process of AD and mild^o^kivejmpairmeiit
Figure imgf000006_0001
due to their role in learning and memory as well as neurogenesis and neuroplasticity, and hence finished the present invention.
The present invention provides a use of benzoic acid salt in the manufacture of a composition for preventing or treating dementia or mild cognitive impairment.
In one aspect of the present application, the benzoic acid salt can be sodium benzoate, potassium benzoate or calcium benzoate, and preferably, the benzoic acid salt is sodium benzoate.
In another aspect of the present application, an effective amount of benzoic acid salt can be 200 milligrams (mg)/day to 2000 mg/day, preferably 500 mg/day to 900 mg/day, and more preferably 750 mg/day.
In a further aspect of the present application, an effective amount of sodium benzoate is 200 mg/day to 2000 mg/day, preferably 500 mg/day to 900 mg/day, and more preferably 750 mg/day.
In one aspect of the present application, the dementia includes early-phase dementia. In one embodiment of the present application, the early-phase dementia includes mild Alzheimer's disease.
In one aspect of the present application, the mild cognitive impairment includes amnestic mild cognitive impairment.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 shows a flow diagram and disposition for two treatment groups.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS The following illustrative embodiments are provided to illustrate the disclosure of the present invention. These and other advantages and effects can be apparently understood by those in the art after reading the disclosure of this specification.
Term definition
. . As used herein, the term "dementia" refers to a group of symptoms affecting intellectual and social abilities severe enough to interfere with daily functioning, including memory loss, language problems, inability to learn or remember new information, etc (78). The term "early-phase dementia" refers to the condition of dementia patients whose CDR (Clinical Dementia Rating) score is not more than 1.
As used herein, the term "Alzheimer's disease (AD)" refers to a kind of progressive mental deteriorative disease that can occur in middle or old age, due to generalized degeneration of the brain, and meet the National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer's Disease and Related Disorders Association criteria. Excessive glutamatergic neurotransmission, particularly through the N-methyl-D-aspartate receptor (NMDAR), leads to neurotoxicity (5,6), which is implicated in the pathophysiology of AD, especially in the late phase. The term "mild Alzheimer's disease" refers to the condition of dementia patients whose CDR (Clinical Dementia Rating) score is 0.5 or 1.
As used herein, the term "mild cognitive impairment (MCI)" refers to a kind of mental deterioration with CDR (Clinical Dementia Rating) score less than 1. The term "amnestic mild cognitive impairment (aMCI)" refers to a kind of MCI in which memory is primarily affected.
As used herein, the term "Clinical Dementia Rating (CDR)" refers to a kind of evaluation system to evaluate the staging severity of dementia. The Clinical Dementia Rating is a five-point scale in which CDR-0 connotes no cognitive impairment, and then the remaining four points are for various stages of dementia: CDR within 0.5-1 = very mild to mild dementia, CDR within 1-2 = mild to moderate, CDR within 2-3 = moderate to severe, CDR > 3 = severe.
Example
The presenijnvention^examined the effieaey and safety of sodium benzoateT^D^^ino acid oxidase (DAAO) inhibitor, for the treatment of amnestic mild cognitive impairment (aMCI) and mild AD.
The inventors conducted a randomized, double-blind, placebo-controlled trial in four major medical centers in Taiwan. Sixty patients with aMCI or mild AD were treated with 250-750 mg/day of sodium benzoate or placebo for 24 weeks. Alzheimer's disease assessment scale-cognitive subscale (ADAS-cog, the primary outcome) and global function (assessed by Clinician Interview Based Impression of Change plus Caregiver Input (CIBIC-plus)) were measured every eight weeks. Additional cognition composite was measured at baseline and endpoint. Participants
Patients were recruited from the outpatient clinics at the Department of Psychiatry and Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Department of Psychiatry, China Medical University Hospital, Taichung, Department of Psychiatry, Taichung Veterans General Hospital, Taichung, and Department of Neurology, Lin-Shin Hospital, Taichung, which are four major medical centers in Taiwan. The study was approved by the institutional IRB at four sites and conducted in accordance with the current revision of the Declaration of Helsinki. Patients were evaluated by research psychiatrists and neurologists after a thorough medical and neurological workup.
Patients were enrolled into this study if they: 1) satisfied NINCDS-ADRDA (National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer's Disease and Related Disorders Association) (39) criteria for probable AD and had a Clinical Dementia Rating (CDR) (40) score of 1, or criteria for aMCI (41) of a presumably degenerative nature defined as subjective memory complaint corroborated by an informant and insufficient global cognitive and functional impairment to meet NINCDS-ADRDA criteria and had a CDR score of 0.5, 2) 50-90 years of age, 3) were physically healthy and had all laboratory assessments (including urine/blood routine, biochemical tests, and electrocardiograph) within normal limits, 4) had a Mini-Mental State Examination (MMSE) (42) score of 17-26, 5) had sufficient education to communicate effectively and were capable of completing the assessments of the study, and 6) agreed to participate in the study and provided informed consent. For patients who had already been on AChEI therapy, AChEI had to be continued for at least three months before enrollment. AChEI dose had to be kept unchanged during the study duration. For patients who had not yet been on AChEI therapy, AChEI or other anti-dementia medication was forbidden during the study duration.
Exclusion criteria included history of significant cerebrovascular disease; Hachinski Ischemic Score >4; major neurological, psychiatric or medical conditions other than AD; substance (including alcohol) abuse or dependence; delusion, hallucination or delirium symptoms; severe visual or hearing loss; and inability to follow protocol.
Study Design
All patients were randomly assigned to receive a 24-week treatment of sodium benzoate or placebo in a double-blind manner. Efficacy and safety were evaluated at baseline and at the ends of weeks 8, 16, and 24. Two hundred and fifty mg of sodium benzoate or placebo were packed with identical capsules provided in coded containers. The dose was started at 250-500 mg/day (250mg once or twice daily) in the first eight weeks, then increased by 250-500 mg/day from the 9th week, and further increased by another 250-500 mg/day from the 17th week of the study if clinically indicated. The inventors decided to apply 250-750 mg/day, considering the older age of the subjects in the present study. Patients were randomized in a cluster of 6 subjects to receive sodium benzoate or placebo in a 1 :1 ratio by an independent investigational pharmacist.
Patients, caregivers and investigators, except the investigational pharmacist, were all blind to the assignment. Patient's medical adherence and safety were closely monitored by caregivers and research physicians and pill-counting by the study staff.
Assessments
The primary outcome was the Alzheimer's disease assessment scale-cognitive subscale (ADAS-cog) (43) measured at weeks 0, 8, 16, and 24. ADAS-cog is the most popular cognitive assessment instrument used in At) clinical trials. It consists of 11 tasks, including word recall, naming, commands, constructional praxis, ideational praxis, orientation, word recognition, instructions remembering, spoken language ability, word-finding difficulty and comprehension. Its scores range from 0 (best) to 70 (worst).
The secondary outcome measurements included the Clinician's Interview-Based Impression of Change plus Caregiver Input (CIBIC-plus) (44) measured at weeks 8, 16 and 24, and the additional cognition composite measured at baseline and endpoint (was measured at the end of each patent's additional cognition composite measurement).
CIBIC-plus is a global assessment of change based on a comprehensive, semi-structured interview which includes caregiver-supplied information. It is a 7-point rating scale ranging from 1-7, where 1 represents markedly improved; 4, no change; and 7, markedly worse.
The additional cognition composite was calculated by the average of the T scores of speed of processing (Category Fluency), working memory (WMS-III, Spatial Span) (45) and verbal learning and memory tests (WMS-III, Word Listing) (45). The raw score of speed of processing, working memory and verbal learning and memory tests was standardized to a T score with a mean of 50 and a standard deviation of 10 for making each test comparative. The additional cognition composite was applied in combination with ADAS-cog to make the cognitive assessment more thorough. Decrease in processing speed has been found to be associated with aging (46, 47). Working memory (48) and verbal learning/memory (49) also decline in patients with AD.
Systemic side effects of treatments were evaluated by means of physical and neurological examinations, laboratory tests including CBC and biochemistry and reviewed by applying the Udvalg for Kliniske Undersogelser (U U) Side-effects Rating Scale (50) at baseline, week 8, 16 and 24.
Clinical ratings were performed by the research psychiatrists and neurologists who were trained and experienced in the rating scales. Inter-rater reliability was analyzed with the ANOVA test. Only raters reaching the intra-class correlation coefficients of > 0.90 during pre-study training were allowed to rate the study patients. To maintain high inter-rater reliability and to prevent rater drift, raters met at least once a quarter for training and reliability re-testing. To minimize inter-rater variability, each individual patient was assessed by the same research psychiatrist or neurologist throughout the trial. Data analysis
Chi-square test (or Fisher's exact test) was used to compare differences of categorical variables and Student's two-sample t-test (or Mann- Whitney U test if the distribution was not normal) for continuous variables between two treatment groups. Mean changes from baseline in repeated-measure assessments (ADAS-cog) were assessed using the generalized estimating equation (GEE) method with treatment, visit and treatment-visit interaction as fixed effects and intercept as the only random effect; baseline value as the covariance. The GEE analyses were performed using the SAS/STAT (SAS Institute, Cary, North Carolina) "PROC
GENMOD" procedure with AR (autoregressive)(7) working correlation structure with the marginal model instead of the mixed effect model. Therapeutic effect sizes (Cohen's d) were used to determine the magnitude of improvement for the continuous variables (51) resulting from sodium benzoate treatment compared with placebo.
Finally, all of the 60 randomized patients completed at least one follow-up, and 50 (90
%) of them completed the 24- week trial (as shown in Figure 1). No imputation for the incomplete data was used for the GEE analysis.
There were no baseline scores for the CIBIC-plus because this was scored as a judgment of change from baseline. Differences in CIBIC-plus scores at week 8, 16, 24 and endpoint between groups were assessed by Student's two-sample t-test (or Mann- Whitney U test if the distribution was not normal).
Fisher's exact test was used to compare differences in the dropout rates between the two groups. Cohen's w was applied for determining the effect size of categorical variables (52).
All data were analyzed by IBM SPSS Statistics (version 18.0; SPSS Inc.) or SAS version 9.3.
All p values for clinical measures were based on two-tailed tests with a significance level of
0.05. Results
Sixty patients were eligible and randomized (as shown in Figure 1). Demographic data, education level, age at illness onset, illness duration, CDR, body mass index (BMI) and AChEI use at baseline were similar between the sodium benzoate group (N = 30) and the placebo group (N = 30) (p > 0.05) (as shown in Table 1). AChEI doses were within the therapeutic range and similar between two groups (as shown in Table 1). Mean dose of sodium benzoate at weeks 8, 16, and 24 were 275.0 ± 76.3, 525.0 ± 100.6, and 716.7 ± 182.6 mg/day, respectively.
Table 1. Baseline-Demographic Gharacteristics of the Placebo or Sodium Benzoate Treatment Group
Treatment Groups
Benzoate (n = 30) Placebo (n = 30) P
Demographic Data
Female, n (%) 18 (60.0) 19 (63.3) 1.0°
Male, n (%) 12 (40.0) 11 (36.7) 1.0
Age, yrs, mean (SD) 70.7 (7.9) 69.7 (9.0) 0.64*
Age at illness onset, yrs, mean (SD) 69.8 (7.1) 68.5 (8.9) 0.54*
Illness duration, months, mean (SD) 14.2 (15.6) 13.6 (17.9) 0.47c
CDR at baseline, n (%) 1.0°
CDR 0.5 15 (50.0) 16 (53.3)
CDR 1 15 (50.0) 14 (46.7)
Education, yrs, mean (SD) 5.9 (4.7) 7.5 (5.2) 0.36c
BMI, mean (SD) 24.6 (4.1) 23.9 (3.4) 0.51*
Patients using AChEIs, n
Total 9 9 1.0°
Donepezil (dose, mean ± SD) 7 (7.9 ± 2.7) 5 (8.0 ± 2.7) 1.0C
Rivastigmine (dose, mean ± SD) 0 (0.0 ± 0.0) 3 (7.5 ± 2.6) NA
Galantamine (dose, mean ± SD) 2 (16.0 ± 0.0) 1 (16.0 ± 0.0) 1.0e
AChEI, acetylcholine esterase inhibitor; BMI, body mass index; CDR, Clinical Dementia Rating; NA, not associated. aFisher's exact test.
^Independent t test.
CMann- Whitney U test The mean ± SD scores for both primary and secondary outcomes, including ADAS -cog, additional cognition composite and CIBIC-plus, of the two groups of patients are shown in Table 2. At week 0 (baseline), there were no significant differences between the two groups in ADAS-cog and additional cognition composite (p = 0.75 and 0.27, respectively).
Table 2. Mean ± SD Scores of Both Primary and Secondary Outcomes
Benzoate Placebo
Scale Estimate" SEM Z p
Mean + SD (n) Mean ± SD (n)
Primary Outcome ADAS-cog
Baseline 15.6 1 7.6 (30) 15.0 + 7.3 (30)
Week 8 11.6 + 6.5 (30) 11.7 ± 8.5 (30) -2.8819 0.9592 -3.00 0.0027
Week 16 9.8 ± 6.2 (29) 12.3 ± 9.1 (26) -1.7582 1.2270 -1.34 0.1519
Week 24 9.7 ± 6.4 (30) 11.3 ± 9.2 (25) -2.7456 1.1845 -2.32 0.0205
Endpoint 9.6 ± 6.2 (30) 12.4 + 9.1 (30) -1.8067 1.1255 -1.61 0.1084
Drug -2.1860 2.2676 -0.96 0.3351
Week 8 x drag -1.0236 1.1491 -0.89 0.3730
Week 16 x drug -4.1835 1.3608 -3.07 0.0021
Week 24 x drug -3.3543 1.3294 -2.52 0.01 16
Endpoint x drug -3.9648 1.3424 -2.95 0.0031 Secondary Outcome
Additional cogn tion
Cohen's d t
composite (T score)
Baseline 48.9 ± 6.6 (26) 51.2 ± 8.4 (27) -1.111 0.272*
Endpoint 50.4 + 6.6 (26) 49.6 ± 8.7 (27) 0.404 0.688*
Difference 1.5 ± 3.1 (26) -1.6 ± 4.8 (27) 0.7826 2.837 0.007*
CIBIC-plus Cohen's d Z P
Week 8 3.4 ± 0.5 (30) 3.5 ± 0.6 (30) 0.2441 -0.869 0.385c
Week 16 3.3 ± 0.6 (30) 3.7 ± 0.7 (26) 0.6637 -2.445 0.015c
Week 24 3.2 ± 0.7 (28) 3.7 ± 0.7 (27) 0.6973 -2.416 0.016c
Endpoint 3.2 ± 0.7 (30) 3.7 ± 0.8 (30) 0.7290 -2.520 0.012c
Results of measures of Alzheimer's Disease Assessment Scale-cognitive subscale (ADAS-cog), additional cognitive tests, and Clinician Interview Based Impression of Change plus Caregiver Input (CIBIC-plus) over the 24-week treatment with generalized estimating equations (GEE) method.
Additional cognition composite: the composite test score of speed of processing, working memory, and verbal learning, and memory.
"Estimate is the coefficient of treatment-visit interaction term in the GEE method multiple linear regression model. A first-order autoregressive
covariance matrix was fit to the within-patient repeated measures. The p values were based on two-tailed tests.
*Independent t test.
'Mann-Whitney U test was used, because the distribution of CIBIC-plus score was not normal.
For the primary outcome, sodium benzoate produced greater improvement in ADAS-cog score than the placebo therapy throughout the study (mean differences from baseline were 3.8, 5.4, 5.9 and 5.9 in the benzoate group, and 2.4, 1.7, 2.7 and 1.7 in the placebo group, at weeks 8, 16, 24 and endpoint; p = 0.3730, 0.0021, 0.0116 and 0.0031, respectively), with effect size of 0.86 at the end of the study (as shown in Table 2). The results were similar when the baseline ADAS-cog score was controlled in the GEE model (as shown in Table SI).
Table SI . Results of measures of ADAS-cog over the 24- week Treatment using generalized estimating equations (GEE) method adjusted for the baseline effect
Scale Benzoate Placebo Estimate* SE Value
Mean ± SD (n) Mean ± SD («)
ADAS-cog
Baseline 15.6 ± 7.6 (30) 15.0 ± 7.3 (30)
-2.6070 0.8801 -2.96 0.0031 Week 8 11.6 ± 6.5 (30) 11.7 ± 8.5 (30)
-1.3145 1.0526 -1.25 0.2118 Week 16 9.8 ± 6.2 (29) 12.3 ± 9.1 (26)
-2.0495 0.9114 -2.25 0.0245 Week 24 9.7 ± 6.4 (28) 11.3 ± 9.2 (25)
-1.9247 0.9507 -2.02 0.0429 Endpoint 9.6 ± 6.2 (30) 12.4 ± 9.1 (30)
0.5135 0.3616 1.42 0.1555 Drag
-1.2397 1.1366 -1.09 0.2754
Week 8 x drug
-4.4638 1.2601 -3.54 0.0004 Week 16 x drug
-3.7523 1.2379 -3.03 0.0024 Week 24 x drug
-3.6255 1.2278 -2.95 0.0031 Endpoint x drug
* Estimate is the coefficient of treatment-visit interaction term in the GEE method's multiple linear regression model. An autoregressive AR(1) covariance matrix was fit to the within-patient repeated measures. P values were based on two-tailed tests.
ADAS-cog: Alzheimer's disease assessment scale-cognitive subscale.
Bolded p values indicate significance. For the secondary outcomes, sodium benzoate was better than placebo in the additional cognition composite at endpoint (p = 0.007, effect size = 0.78). Benzoate treatment also produced greater improvement in CIBIC-plus score than placebo therapy at week 16 (p - 0.015), week 24 (p = 0.016) and endpoint (p = 0.012, effect size = 0.73 at endpoint) (as shown in Table 2).
The dropout rate (3.3%) of sodium benzoate group tended to be lower than that (16.7%) of the placebo group, yet insignificantly (p = 0.195).
For subgroup analysis, we further examined efficacy of sodium benzoate vs. placebo in
CDR 0.5 and CDR 1 subgroups. For ADAS-cog, sodium benzoate produced greater improvement than placebo therapy at week 16, 24 and endpoint (p = 0.0151, 0.0387 and 0.0092, respectively) in the CDR 1 subgroup. However, sodium benzoate was not superior to the placebo therapy in the CDR 0.5 subgroup throughout the study (p > 0.05) (as shown in
Table 3).
Although ADAS-cog is widely used in AD clinical trials, it may be less sensitive for MCI (67). One of the strategies to improve the detection of responsiveness for MCI is to add additional cognitive tests. People with MCI have been found to be impaired in neuropsychological functions (68) such as speed of processing (69), working memory (70) and verbal learning and memory (71). In the aMCI subgroup of the present invention, sodium benzoate showed borderline significance in improving the additional cognition composite, consisting of speed of processing, working memory, and verbal learning/memory, but not in ADAS-cog score. Our result echoes the suggestion that additional neuropsychological tests which are more sensitive to subtle deficits should also be applied in the trials for MCI.
Table 3. Results of Measures of ADAS-cog Over 24- Week Treatment With GEE Method in Subgroups
Benzoate Placebo
Scale Estimate" SEM Z P
Mean ± SD (n) Mean ± SD (n)
CDR 0.5
Baseline 13.8 + 6.1 (15) 11.9 ± 5.4 (16) Reference 0.3560
Week 8 x drug 10.4 ± 6.0 (15) 8.3 + 4.0 (16) 0.3213 0.9690 0.33 0.7402
Week 16 x drug 9.1 + 6.1 (15) 10.2 ± 5.6 (15) -3.2694 1.9222 -1.70 0.0890
Week 24 x drug 9.5 + 6.2 (14) 8.7 ± 5.0 (14) -1.8480 1.7834 -1.04 0.3001
Endpoint x drug 9.2 ± 6.1 (15) 9.8 ± 5.9 (16) -2.3444 1.9664 -1.19 0.2332
CDR 1
Baseline 17.1 ± 8.6 (15) 16.7 ± 8.2 (14) Reference 0.8910
Week 8 x drug 13.0 ± 7.1 (15) 15.6 ± 10.6 (14) -2.5727 2.0872 -1.23 0.2177
Week 16 x drug 10.6 ± 6.4 (14) 15.2 ± 12.2 (11) -55.0788 2.0897 -2.43 0.0151
Week 24 x drug 9.9 ± 6.8 (14) 14.5 ± 12.3 (11) -4.6262 2.2375 -2.07 0.0387
Endpoint x drug 10.0 ± 6.6 (15) 15.4 + 11.3 (14) -5.4755 2.1031 -2.60 0.0092
Abbreviations are the same as those in Tables 1 and 2.
"Estimate is the coefficient of treatment-visit interaction term in the GEE method multiple linear regression model. A first-order autoregressive covariance matrix was fit to the within-patient repeated measures. The p values were based on two-tailed tests.
Sodium benzoate showed better efficacy in the CDR 1 subgroup (p = 0.041) and borderline significance in the CDR 0.5 subgroup (p = 0.063) in improving the additional cognition composite (as shown in Table 4). For CIBIC-plus, sodium berizoate produced greater improvement than placebo therapy at week 24 and endpoint (p = 0.040 and 0.018, respectively) in the CDR 1 subgroup, but not in the CDR 0.5 subgroup (as shown in Table 5).
Sodium benzoate also did not improve CIBIC-plus score in the aMCI subgroup. A possible explanation is a ceiling effect that functional impairment is minimal in the MCI individuals, thereby restricting the space for further improvement.
Table 4. Results of Measures of Additional Cognition Composite Over 24-Week Treatment with Independent t Test in Subgroups
Benzoate Placebo
Scale Mean ± SD (n) Mean ± SD (n) Cohen's d t
CDR 0.5
Baseline 49.0 ± 6.9 (14) 52.8 ± 7.6 (15) -1.395 .174
Endpoint 50.5 ± 7.1 (14) 50.8 ± 8.5 (15) -.108 .915
Difference 1.5 ± 3.5 (14) -1.9 ± 5.8 (15) .7098 1.939 .063
CDR 1
Baseline 48.8 ± 6.6 (12) 49.3 ± 9.3 (12) .150 .882
Endpoint 50.3 ± 6.1 (12) 48.0 ± 9.0 (12) .742 .466
Difference 1.6 ± 2.8 (12) -1.3 ± 3.5 (12) .9150 2.176 .041 The p values were based on two-tailed tests. Additional cognition composite, the composite test score of speed of processing, working memory, and verbal learning and memory.CDR, Clinical Dementia Rating.
Table 5. Results of Clinical Measures of CIBIC-Plus Over 24-Week Treatment with
Mann- Whitney U Test in Subgroups
Benzoate Placebo
Scale Mean ± SD («) Mean i SD («) Cohen's d Z P
CDR 0.5
Week 8 3.5 ± .5 (15) 3.4 ± .5 (16) .1771 -.508 .611
Week 16 3.3 ± .5 (15) 3.7 ± .8 (15) -.6042 -1.720 .085
Week 24 3.3 ± .5 (14) 3.6 ± .6 (16) -.4867 -1.260 .208
Endpoint 3.3 ± .5 (15) 3.6 ± .6 (16) -.4086 -1.029 .303
CDR 1
Week 8 3.3 ± .5 (15) 3.6 ± .6 (14) -.6697 -1.719 .086
Week 16 3.2 ± .7 (15) 3.6 ± .5 (11) -.7374 -1.678 .093
Week 24 3.1 ± .8 (14) 3.8 ± .9 (11) -.8805 -2.052 .040
Endpoint 3.1 ± .8 (15) 3.9 ± .9 (14) -.9494 -2.370 .018 The p values were based on two-tailed tests. Mann- Whitney U test was used because the distribution of CIBIC-plus score was not normal.
CD , Clinical Dementia Rating;
CIBIC-plus, Clinician Interview Based Impression of Change plus Caregiver Input.
It is critical to identify and treat AD as early as possible, potentially to arrest its progression (53). The present invention is the first to apply a DAAO inhibitor, sodium benzoate herein, as a novel treatment for the early stage of cognitive decline. The result showed that sodium benzoate had better efficacy than placebo in improving ADAS-cog score, additional cognition composite (consisting of speed of processing, working memory, verbal learning and memory) and global function in all subjects as a whole. Subgroup comparisons found that sodium benzoate was beneficial for all outcome measures among patients with mild AD. In the aMCI subgroup, sodium benzoate showed borderline significance in improving the cognition composite. Moreover, sodium benzoate also demonstrated favorable safety profiles.
As to the dosing strategy, sodium benzoate provided better efficacy than placebo at week 16 and week 24, with the mean dose of 525 mg/day and 716 mg/day respectively, possibly implying that sodium benzoate at 500-750 mg/day is more effective than 250 mg/day. Another possibility is that longer sodium benzoate treatment duration yields better treatment response.
AChEIs are commonly used for the treatment of AD (57, 58), but are not recommended for the treatment of MCI due to weak beneficial effects and risk of side effects (59, 60). The consensus statement from the British Association for Psychopharmacology concludes that neither AChEIs nor memantine is effective in treating MCI (61). Other compounds commonly used for the treatment of MCI, such as vitamin E (62), folic acid (63), omega-3 fatty acid (64), piracetam (65) and Ginkgo biloba (66), also failed to show convincing evidence for a cognitive enhancing effect. Sodium benzoate is generally safe and its efficacy for aMCI reached a trend of improvement in the current small-sized study.
Very high levels of DAAO are detected in the cerebellum of adult brain whereas the activity of DAAO is low in the forebrain such as prefrontal cortex and hippocampus despite robust expression (75, 76). The cellular localization and function of DAAO are likely different between forebrain and cerebellum: it is glial in the cerebellum, but mainly neuronal in the cerebral cortex. However, the effect of DAAO inhibitors on forebrain D-serine level is inconsistent. Most DAAO inhibitors can, while some inhibitors may not cause a measurable increase in D-serine in the forebrain as observed in the cerebellum (77). Nevertheless, cerebellum is involved in cognition. Sodium benzoate may exert its procognitive effects by not only cerebral but also cerebellar mechanism.
The present invention suggests that sodium benzoate, a DAAO inhibitor, is beneficial for cognitive and overall function in patients with early-phase AD and potentially beneficial for aMCI. The use of sodium benzoate for early AD and aMCI will bring hope for the growing aging population with cognitive decline. The results of the present application suggest that benzoate treatment for early-phase dementia may be due to activation of neurogenesis and anti-apoptosis.
Adverse Effects
Both sodium benzoate and placebo were well tolerated. Only one patient in the placebo group reported dizziness at week 16. The side effect was mild and not warranting medical treatment. There was no reported side effect in the sodium benzoate group assessed by the UKU Side-effects Rating Scale at all visits. No dropout was due to side effect.
The routine blood cell count and chemistry were all within the normal ranges and remained unchanged after-treatment (data not shown). This work was supported by the National Science Council, Taiwan (NSC 99-3114-B-182A-003, NSC Ϊ01-2314-Β-182Α-073-ΜΥ2, and NSC-101 -2325-B-039-009), Taiwan Department of Health Clinical Trial and Research Center of Excellence (DOH102-TD-B-111-004), and China Medical University Hospital, Taiwan (CMU 101-AWARD-13, DMR-99-153).
The foregoing descriptions of the detailed embodiments are only illustrated to disclose the principle and functions of the present invention and do not restrict the scope of the present invention. It should be understood to those in the art that all modifications and variations according to the spirit and principle in the disclosure of the present invention should fall within the scope of the appended claims. It is intended that the specification and examples are considered as exemplary only, with a true scope of the invention being indicated by the following claims.
The references listed below in the application are each incorporated by reference as if they were incorporated individually.
1. Budd D, Burns LC, Guo Z, L'ltalien G, Lapuerta P (2011): Impact of early intervention and disease modification in patients with predementia Alzheimer's disease: a Markov model simulation. ClinicoEconomics and outcomes research : CEOR. 3:189-195.
2. Loy C, Schneider L (2006): Galantamine for Alzheimer's disease and mild cognitive impairment. Cochrane Database Syst Rev. CDOOl 747.
3. Raschetti R, Albanese E, Vanacore N, Maggini M (2007): Cholinesterase inhibitors in mild cognitive impairment: a systematic review of randomised trials. PLoS medicine. 4:e338.
4. Birks J, Flicker L (2006): Donepezil for mild cognitive impairment. Cochrane Database
Figure imgf000023_0001
5. Lipton SA, Rosenberg PA (1994): Excitatory amino acids as a final common pathway for neurologic disorders. NEngl J Med. 330:613-622. 6. alia LV, alia SK, Salter MW (2008): NMDA receptors in clinical neurology:
excitatory times ahead. Lancet Neurol. 7:742-755.
7. Choi DW (1992): Excitotoxic cell death. Journal of neurobiology. 23:1261-1276.
8. Scarpini E, Scheltens P, Feldman H (2003): Treatment of Alzheimer's disease: current status and new perspectives. Lancet neurology. 2:539-547.
9. Gardoni F, Mauceri D, Malinverno M, Polli F, Costa C, Tozzi A, et al. (2009): Decreased NR2B subunit synaptic levels cause impaired long-term potentiation but not long-term depression. JNeurosci. 29:669-677.
10. Pallas M, Camins A (2006): Molecular and Biochemical Features in Alzheimer disease. Curr Pharm Des. 12:4389-4408.
11. Reisberg B, Doody R, Stoffler B, Schmitt F, Ferris S, Mobius HJ (2003): Memantine in Moderate-to-Severe Alzheimer Disease. N Engl J Med. 348:1333-1341.
12. Schneider LS, Dagerman S, Higgins JP, McShane R (2011): Lack of evidence for the efficacy of memantine in mild Alzheimer disease. Arch Neurol. 68:991-998.
13. Yoon WJ, Won SJ, Ryu BR, Gwag BJ (2003): Blockade of ionotropic glutamate receptors produces neuronal apoptosis through the Bax-cytochrome C-caspase pathway: the causative role of Ca2+ deficiency. J Neurochem. 85:525-533.
14. Rowland LM, Astur RS, Jung RE, Bustillo JR, Lauriello J, Yeo RA (2005): Selective cognitive impairments associated with NMDA receptor blockade in humans.
Neuropsychopharmacology. 30:633-639.
15. Mattson MP (2008): Glutamate and neurotrophic factors in neuronal plasticity and disease. Ann NY Acad Sci. 1144:97-112.
16. Tilleux S, Hermans E (2007): Neuroinflammation and regulation of glial glutamate uptake in neurological disorders. JNeurosci Res. 85:2059-2070.
17. Olney JW, Farber NB (1995): Glutamate receptor dysfunction and schizophrenia. Arch Gen Psychiatry. 52:998-1007.
18. Segovia G, Porras A, Del Arco A, Mora F (2001): Glutamatergic neurotransmission in aging: a critical perspective. Mech Ageing Dev. 122:1-29.
19. Procter AW, Stirling JM, Stratmann GC, Cross AJ, Bowen DM (1989): Loss of glycine-dependent radioligand binding to the N-methyl-D-aspartate-phencyclidine receptor complex in patients with Alzheimer's disease. Neurosci Lett. 101 :62-66.
20. Procter AW, Wong EH, Stratmann GC, Lowe SL, Bowen DM (1989): Reduced glycine stimulation of [3HJMK-801 binding in Alzheimer's disease. J Neurochem. 53 :698-704.
21. Chessell IP, Procter AW, Francis PT, Bowen DM (1991): D-cycloserine, a putative cognitive enhancer, facilitates activation of the N-methyl-D-aspartate receptor-ionophore complex in Alzheimer brain. Brain Res. 565:345-348.
22. Tsai GE, Falk WE, Gunther J, Coyle JT (1999): Improved cognition in Alzheimer's disease with short-term D-cycloserine treatment. Am J Psychiatry. 156:467-469.
23. Lane HY, Lin CH, Green MF, Hellemann G, Huang CC, Chen PW, et al. (2013): A Randomized, Double-Blind, Placebo-Controlled Add-on Treatment of Benzoate, a D-Amino Acid Oxidase Inhibitor, for Schizophrenia. JAMA Psychiatry. 70: 1267-1275.
24. Fukui K, Miyake Y (1992): Molecular cloning and chromosomal localization of a human gene encoding D-amino-acid oxidase. The Journal of biological chemistry.
267:18631-18638.
25. Vanoni MA, Cosma A, Mazzeo D, Mattevi A, Todone F, Curti B (1997): Limited proteolysis and X-ray crystallography reveal the origin of substrate specificity and of the rate-limiting product release during oxidation of D-amino acids catalyzed by mammalian D-amino acid oxidase. Biochemistry. 36:5624-5632.
26. Sasabe J, Miyoshi Y, Suzuki M, Mita M, Konno R, Matsuoka M, et al. (2012): D-amino acid oxidase controls motoneuron degeneration through D-serine. Proc Natl Acad Sci USA. 109:627-632.
27. Esposito S, Pristera A, Maresca G, Cavallaro S, Felsani A, Florenzano F, et al. (2012): Contribution of serine racemase/d-serine pathway to neuronal apoptosis. Aging Cell
11 :588-598.
28. Huang X, Kong H, Tang M, Lu M, Ding JH, Hu G (2012): D-Serine regulates proliferation and neuronal differentiation of neural stem cells from postnatal mouse forebrain. CNS Neurosci Ther. 18:4-13.
29. Smith SM, Uslaner JM, Hutson PH (2010): The Therapeutic Potential of D- Amino Acid
Oxidase (DAAO) Inhibitors. The open medicinal chemistry journal. 4:3-9.
30. WHO-Concise International Chemical Assessment Document No.26. WHO G, 2000 http://www. inchem. org/documents/cicads/cicads/cicad26. htm.
31. World Health Organization (2000): Concise International Chemical Assessment.
Document No. 26. Geneva: World Health Organization. Available at:
http://www.who.int/ipcs/publications/cicad/cicad26_rev_l.pdf..
32. Smith SM, Uslaner JM, Yao L, Mullins CM, Surles NO, Huszar SL, et al. (2009): The behavioral and neurochemical effects of a novel D-amino acid oxidase inhibitor compound 8
[4H-thieno [3,2-b]pyrrole-5-carboxylic aeid] and D-serine. The Journal of pharmacology and experimental therapeutics. 328:921-930.
33. Adage T, Trillat AC, Quattropani A, Perrin D, Cavarec L, Shaw J, et al. (2008): In vitro and in vivo pharmacological profile of AS057278, a selective d-amino acid oxidase inhibitor with potential anti-psychotic properties. Eur Neuropsychopharmacol. 18:200-214.
34. Hashimoto K, Fujita Y, Horio M, Kunitachi S, Iyo M, Ferraris D, et al. (2009):
Co-administration of a D-amino acid oxidase inhibitor potentiates the efficacy of D-serine in attenuating prepulse inhibition deficits after administration of dizocilpine. Biol Psychiatry. 65:1103-1106. 35. Zhao WJ, Gao ZY, Wei H, Nie HZ, Zhao Q, Zhou XJ, et al. (2010): Spinal D-amino acid oxidase contributes to neuropathic pain in rats. The Journal of pharmacology and
experimental therapeutics. 332:248-254.
36. Gong N, Gao ZY, Wang YC, Li XY, Huang JL, Hashimoto K, et al. (2011 ): A series of D-amino acid oxidase inhibitors specifically prevents and reverses formalin-induced tonic pain in rats. The Journal of pharmacology and experimental therapeutics. 336:282-293.
37. Park HK, Shishido Y, Ichise-Shishido S, Kawazoe T, Ono K, Iwana S, et al. (2006): Potential role for astroglial D-amino acid oxidase in extracellular D-serine metabolism and cytotoxicity. Journal of biochemistry. 139:295-304.
38. Batshaw ML, Hyman SL, Coyle JT, Robinson MB, Qureshi IA, Mellits ED, et al. (1988): Effect of sodium benzoate and sodium phenylacetate on brain serotonin turnover in the ornithine transcarbamylase-deficient sparse-fur mouse. Pediatric research. 23:368-374.
39. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM (1984):
Clinical diagnosis of Alzheimer's disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer's Disease. Neurology. 34:939-944.
40. Morris JC (1993): The Clinical Dementia Rating (CDR): current version and scoring rules. Neurology. 43:2412-2414.
41. Lu PH, Edland SD, Teng E, Tingus K, Petersen RC, Cummings JL (2009): Donepezil delays progression to AD in MCI subjects with depressive symptoms. Neurology.
72:2115-2121.
42. Folstein MF, Folstein SE, McHugh PR (1975): "Mini-mental state". A practical method for grading the cognitive state of patients for the clinician. Journal of psychiatric research. 12:189-198.
43. Rosen WQ Mohs RC, Davis KL ( 1984) : A new rating scale for Alzheimer's disease. Am J Psychiatry. 141 :1356-1364.
44. Schneider LS, Olin JT, Doody RS, Clark CM, Morris JC, Reisberg B, et al. (1997): Validity and reliability of the Alzheimer's Disease Cooperative Study-Clinical Global Impression -of Change^The Alzheimer's Disease Cooperative Study Alzh^imeYDis Assoc^ Disord. 11 Suppl 2:S22-32.
45. Wechsler D (1997): Wechsler Memory Scale, 3rd ed. Psychological Association, San Antonio, TX Psychological Association.
46. Salthouse TA (1996): The processing-speed theory of adult age differences in cognition. Psychological review. 103:403-428.
47. Habekost T, Vogel A, Rostrup E, Bundesen C, Kyllingsbaek S, Garde E, et al. (2013): Visual processing speed in old age. Scandinavian journal of psychology. 54:89794.
48. Carlesimo GA, Mauri M, Graceffa AM, Fadda L, Loasses A, Lorusso S, et al. (1998): Memory performances in young, elderly, and very old healthy individuals versus patients with Alzheimer's disease: evidence for discontinuity between normal and pathological aging. Journal of clinical and experimental neuropsychology. 20:14-29.
49. Chen P, Ratcliff G, Belle SH, Cauley JA, DeKosky ST, Ganguli M (2001): Patterns of cognitive decline in presymptomatic Alzheimer disease: a prospective community study. Arch Gen Psychiatry. 58:853-858.
50. Lingjaerde O, Ahlfors UG, Bech P, Dencker S J, Elgen K (1987): The U U side effect rating scale. A new comprehensive rating scale for psychotropic drugs and a cross-sectional study of side effects in neuroleptic-treated patients. Acta psychiatrica Scandinavica
Supplementum. 334:1-100.
51. Rosenthal R RR (1991): Essentials of behavioral research: Methods and data analysis. 2nd ed. New York: McGraw Hill.
52. Cunningham JB M-GE (2007): Power, effect and sample size using GPower: practical issues for researchers and members of research ethics committees. Evidence Based Midwifery 5:132-136.
53. Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S, Fagan AM, et al. (2011): Toward defining the preclinical stages of Alzheimer's disease: recommendations from the National Institute on Aging- Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimer's & dementia : the journal of the Alzheimer's Association. 7:280-292.
54. Huang YJ, Lin CH, Lane HY, Tsai GE (2012): NMDA Neurotransmission Dysfunction in Behavioral and Psychological Symptoms of Alzheimer's Disease. Current
neuropharmacology. 10:272-285.
55. Riederer P, Hoyer S (2006): From benefit to damage. Glutamate and advanced glycation end products in Alzheimer brain. J Neural Transm. 113:1671-1677.
56. Hashimoto K, Fukushima T, Shimizu E, Okada S, Komatsu N, Okamura N, et al. (2004): Possible role of D-serine in the pathophysiology of Alzheimer's disease. Prog
Neuropsychopharmacol Biol Psychiatry. 28:385-388.
57. Birks J (2006): Cholinesterase inhibitors for Alzheimer's disease. Cochrane Database SystRev.CD005593.
58. Burns A, O'Brien J, Auriacombe S, Ballard C, Broich K, Bullock R, et al. (2006):
Clinical practice with anti-dementia drugs: a consensus statement from British Association for Psychopharmacology. J Psychopharmacol. 20:732-755.
59. Fellgiebel A (2007): [Alzheimer drugs for mild cognitive impairment].
Neuropsychiatrie : Klinik, Diagnostik, Therapie und Rehabilitation : Organ der Gesellschafi Osterreichischer Nervenarzte und Psychiater. 21 :230-233.
60. Russ TC, Morling JR (2012): Cholinesterase inhibitors for mild cognitive impairment. Cochrane Database Syst Rev. 9:CD009132. 61. O'Brien JT, Bums A (2011): Clinical practice with anti-dementia drugs: a revised (second) consensus statement from the British Association for Psychopharmacology. J Psychopharmacol. 25:997-1019.
62. Farina N, Isaac MG, Clark AR, Rusted J, Tabet N (2012): Vitamin E for Alzheimer's dementia and mild cognitive impairment. Cochrane Database Syst Rev. 11 :CD002854.
63. Malouf R, Grimley Evans J (2008): Folic acid with or without vitamin B12 for the prevention and treatment of healthy elderly and demented people. Cochrane Database Syst Jtev.CD004514.
64. Sydenham E, Dangour AD, Lim WS (2012): Omega 3 fatty acid for the prevention of cognitive decline and dementia. Cochrane Database Syst Rev. 6:CD005379.
65. Flicker L, Grimley Evans G (2001): Piracetam for dementia or cognitive impairment. Cochrane Database Syst Rev.CDOOlOll.
66. Birks J, Grimley Evans J (2009): Ginkgo biloba for cognitive impairment and dementia. Cochrane Database Syst Rev. CD003120.
67. Skinner J, Carvalho JO, Potter GG, Thames A, Zelinski E, Crane PK, et al. (2012): The Alzheimer's Disease Assessment Scale-Cognitive-Plus (ADAS-Cog-Plus): an expansion of the ADAS-Cog to improve responsiveness in MCI. Brain imaging and behavior. 6:489-501. 68. Salmon DP (2012): Neuropsychological features of mild cognitive impairment and preclinical Alzheimer's disease. Current topics in behavioral neurosciences. 10:187-212. 69. Price SE, Kinsella GJ, Ong B, Storey E, Mullaly E, Phillips M, et al. (2012): Semantic verbal fluency strategies in amnestic mild cognitive impairment. Neuropsychology.
26:490-497.
70. Rios C, Pascual LF, Santos S, Lopez E, Fernandez T, Navas I, et al. (2001): [Working memory and complex activities of everyday life in the initial stages of Alzheimer's disease]. Revista de neurologia. 33:719-722. 71. Espinosa A, Alegret M, Valero S, Vinyes-Junque G, Hernandez I, Mauleon A, et al. (2013): A longitudinal follow-up of 550 mild cognitive impairment patients: evidence for large conversion to dementia rates and detection of major risk factors involved. JAlzheimers Dis. 34:769-780.
72. Albert M,. Blacker D, Moss MB, Tanzi R, McArdle JJ (2007): Longitudinal change in cognitive performance among individuals with mild cognitive impairment. Neuropsychology. 21:158-169.
73. Perneczky R, Pohl C, Sorg C, Hartmann J, Komossa K, Alexopoulos P, et al. (2006): Complex activities of daily living in mild cognitive impairment: conceptual and diagnostic issues. Age and ageing. 35:240-245.
74. Lai CH, Lane HY, Tsai GE (2012): Clinical and cerebral volumetric effects of sodium benzoate, a D-amino acid oxidase inhibitor, in a drug-naive patient with major depression. Biol Psychiatry. 71:e9-el0.
75. Kapoor R, Lim KS, Cheng A, Garrick T, Kapoor V (2006): Preliminary evidence for a link between schizophrenia and NMD A-glycine site receptor ligand metabolic enzymes, d-amino acid oxidase (DAAO) and kynurenine aminotransferase- 1 (KAT-1). Brain Res.
1106:205-210.
76. Verrall L, Walker M, Rawlings N, Benzel I, Kew JN, Harrison PJ, et al. (2007):
d- Amino acid oxidase and serine racemase in human brain: normal distribution and altered expression in schizophrenia. The European journal of neuroscience. 26: 1657-1669.
77. Strick CA, Li C, Scott L, Harvey B, Hajos M, Steyn SJ, et al. (2011): Modulation of NMDA receptor function by inhibition of D-amino acid oxidase in rodent brain.
Neuropharmacology. 61:1001-1015.
78. http://www.mayoclinic.org/diseases-conditions/alzheimers-disease/expert-blog/ dementia-definitions/bgp-20055922

Claims

What is claimed is:
I . A use of benzoic acid salt in the manufacture of a composition for preventing or treating dementia or mild cognitive impairment.
2. The use according to claim 1, wherein the benzoic acid salt is sodium benzoate, potassium benzoate or calcium benzoate.
3. The use according to claim 1 , wherein the benzoic acid salt is sodium benzoate.
4. The use according to claim 1 , wherein the dementia is early-phase dementia.
5. The use according to claim 4, wherein the early-phase dementia comprises mild Alzheimer's disease.
6. The use according to claim 1, wherein the mild cognitive impairment comprises amnestic mild cognitive impairment.
7. The use according to claim 1, wherein an effective amount of benzoic acid salt is 200 milligrams (mg)/day to 2000 mg/day.
8. The use according to claim I, wherein an effective amount of benzoic acid salt is
500 mg/day to 900 mg/day.
9. The use according to claim 1 , wherein an effective amount of benzoic acid salt is 750 mg/day.
10. The use according to claim 3, wherein an effective amount of sodium benzoate is 200 mg/day to 2000 mg/day.
I I . The use according to claim 3, wherein an effective amount of benzoic acid salt is 500 mg/day to 900 mg/day.
12. The use according to claim 3, wherein an effective amount of sodium benzoate is 750 mg/day.
PCT/SG2014/000140 2014-03-24 2014-03-24 Use of benzoic acid salt in the manufactue of a composition for preventing or treating dementia or mild cognitive impairment WO2015147742A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
SG11201507188QA SG11201507188QA (en) 2014-03-24 2014-03-24 Use of benzoic acid salt in the manufacture of a composition for preventing or treating dementia or mild cognitive impairment
KR1020177024853A KR102162073B1 (en) 2014-03-24 2014-03-24 Use of benzoic acid salt in the manufacture of a composition for preventing or treating dementia or mild cognitive impairment
KR1020167003184A KR20160029837A (en) 2014-03-24 2014-03-24 Use of benzoic acid salt in the manufacture of a composition for preventing or treating dementia or mild cognitive impairment
PCT/SG2014/000140 WO2015147742A1 (en) 2014-03-24 2014-03-24 Use of benzoic acid salt in the manufactue of a composition for preventing or treating dementia or mild cognitive impairment
DE212014000063.7U DE212014000063U1 (en) 2014-03-24 2014-03-24 Use of benzoic acid salt in the manufacture of a composition for the prevention or treatment of dementia or mild cognitive impairment
AU2014386718A AU2014386718B8 (en) 2014-03-24 2014-03-24 Use of benzoic acid salt in the manufactue of a composition for preventing or treating dementia or mild cognitive impairment
JP2016510654A JP2016517866A (en) 2014-03-24 2014-03-24 Use of benzoate for the manufacture of a composition for preventing or treating dementia or mild cognitive impairment
CA2902498A CA2902498C (en) 2014-03-24 2014-03-24 Use of benzoic acid salt in the manufactue of a composition for preventing or treating dementia or mild cognitive impairment

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/SG2014/000140 WO2015147742A1 (en) 2014-03-24 2014-03-24 Use of benzoic acid salt in the manufactue of a composition for preventing or treating dementia or mild cognitive impairment

Publications (1)

Publication Number Publication Date
WO2015147742A1 true WO2015147742A1 (en) 2015-10-01

Family

ID=54196083

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SG2014/000140 WO2015147742A1 (en) 2014-03-24 2014-03-24 Use of benzoic acid salt in the manufactue of a composition for preventing or treating dementia or mild cognitive impairment

Country Status (7)

Country Link
JP (1) JP2016517866A (en)
KR (2) KR102162073B1 (en)
AU (1) AU2014386718B8 (en)
CA (1) CA2902498C (en)
DE (1) DE212014000063U1 (en)
SG (1) SG11201507188QA (en)
WO (1) WO2015147742A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018160055A1 (en) * 2017-03-03 2018-09-07 Excelsior Pharmatech Labs Method for preventing or treating autism spectrum disorders by benzoic acid salt
WO2019103597A1 (en) 2017-11-22 2019-05-31 Excelsior Pharmatech Labs Benzoic acid or a salt and derivative thereof for use in preventing or treating depression
IL266210A (en) * 2016-10-24 2019-06-30 Syneurx Int Taiwan Corp Polymorphic forms of sodium benzoate and uses thereof
JP2019518756A (en) * 2016-06-13 2019-07-04 シニュークス インターナショナル(タイワン)コーポレイション Sodium benzoate co-crystal and its use
JP2019518757A (en) * 2016-06-13 2019-07-04 シニュークス インターナショナル(タイワン)コーポレイション Lithium benzoate co-crystal and its use
US11369579B2 (en) 2016-10-24 2022-06-28 Syneurx International (Taiwan) Corp. Polymorphic forms of sodium benzoate and uses thereof
RU2782627C2 (en) * 2016-10-24 2022-10-31 Сайньюрекс Интернэшнл (Тайвань) Корп. Polymorphous forms of sodium benzoate and their use

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010085452A1 (en) * 2009-01-20 2010-07-29 Los Angeles Biomedical Research Institute At Harbor - Ucla Medical Center Sorbic and benzoic acid and derivatives thereof enhance the activity of a neuropharmaceutical

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8601915D0 (en) * 1986-01-27 1986-03-05 Efamol Ltd Pharmaceutical compositions
CN102010382A (en) * 2003-12-29 2011-04-13 塞普拉科公司 Benzo[d]isoxazol-3-ol daao inhibitors
GB201111704D0 (en) * 2011-07-07 2011-08-24 Takeda Pharmaceutical Novel compounds
JP6169492B2 (en) * 2011-11-15 2017-07-26 武田薬品工業株式会社 Dihydroxy aromatic heterocyclic compounds

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010085452A1 (en) * 2009-01-20 2010-07-29 Los Angeles Biomedical Research Institute At Harbor - Ucla Medical Center Sorbic and benzoic acid and derivatives thereof enhance the activity of a neuropharmaceutical

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LIN, C-H. ET AL.: "Benzoate, A D-Amino Acid Oxidase Inhibitor, for the Treatment of Early-Phase Alzheimer Disease: A Randomized, Double-Blind, Placebo-Controlled Trial''.", BIOLOGICAL PSYCHIATRY, vol. 75, no. 9, 2014, pages 678 - 685, XP028867799, [retrieved on 20130925] *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11739046B2 (en) 2016-06-13 2023-08-29 Syneurx International (Taiwan) Corp. Co-crystals of lithium benzoate and uses thereof
JP2019518756A (en) * 2016-06-13 2019-07-04 シニュークス インターナショナル(タイワン)コーポレイション Sodium benzoate co-crystal and its use
JP2019518757A (en) * 2016-06-13 2019-07-04 シニュークス インターナショナル(タイワン)コーポレイション Lithium benzoate co-crystal and its use
US11369579B2 (en) 2016-10-24 2022-06-28 Syneurx International (Taiwan) Corp. Polymorphic forms of sodium benzoate and uses thereof
IL266210A (en) * 2016-10-24 2019-06-30 Syneurx Int Taiwan Corp Polymorphic forms of sodium benzoate and uses thereof
RU2782627C2 (en) * 2016-10-24 2022-10-31 Сайньюрекс Интернэшнл (Тайвань) Корп. Polymorphous forms of sodium benzoate and their use
EP3529229A4 (en) * 2016-10-24 2020-09-30 Syneurx International (Taiwan) Corp. Polymorphic forms of sodium benzoate and uses thereof
AU2018227291B2 (en) * 2017-03-03 2021-02-11 Excelsior Pharmatech Labs Method for preventing or treating autism spectrum disorders by benzoic acid salt
WO2018160055A1 (en) * 2017-03-03 2018-09-07 Excelsior Pharmatech Labs Method for preventing or treating autism spectrum disorders by benzoic acid salt
US10987327B2 (en) 2017-03-03 2021-04-27 Excelsior Pharmatech Labs Method for preventing or treating autism spectrum disorders by benzoic acid salt
JP2021504471A (en) * 2017-11-22 2021-02-15 林 潔欣LIN, Chieh−Hsin Benzoic acid or its salts and derivatives to prevent or treat depression
JP7034314B2 (en) 2017-11-22 2022-03-11 潔欣 林 Benzoic acid or its salts and derivatives to prevent or treat depression
JP2022071083A (en) * 2017-11-22 2022-05-13 潔欣 林 Benzoic acid or salts and derivatives thereof for preventing or treating depression
AU2018371628B2 (en) * 2017-11-22 2021-08-12 Excelsior Pharmatech Labs Benzoic acid or a salt and derivative thereof for use in preventing or treating depression
CN111491627A (en) * 2017-11-22 2020-08-04 林洁欣 Use of benzoic acid or salts and derivatives thereof for preventing or treating melancholia
JP7312286B2 (en) 2017-11-22 2023-07-20 潔欣 林 Benzoic acid or its salts and derivatives for preventing or treating depression
WO2019103597A1 (en) 2017-11-22 2019-05-31 Excelsior Pharmatech Labs Benzoic acid or a salt and derivative thereof for use in preventing or treating depression
US11826326B2 (en) 2017-11-22 2023-11-28 Excelsior Pharmatech Labs Benzoic acid or a salt and derivative thereof for use in preventing or treating depression

Also Published As

Publication number Publication date
AU2014386718A8 (en) 2017-12-07
DE212014000063U1 (en) 2015-10-12
KR20170104658A (en) 2017-09-15
CA2902498F (en) 2015-09-24
AU2014386718A1 (en) 2015-10-08
AU2014386718B8 (en) 2017-12-07
CA2902498C (en) 2017-03-28
KR102162073B1 (en) 2020-10-07
SG11201507188QA (en) 2015-10-29
AU2014386718B2 (en) 2017-07-20
KR20160029837A (en) 2016-03-15
CA2902498A1 (en) 2015-09-24
JP2016517866A (en) 2016-06-20

Similar Documents

Publication Publication Date Title
Lin et al. Benzoate, a D-amino acid oxidase inhibitor, for the treatment of early-phase Alzheimer disease: a randomized, double-blind, placebo-controlled trial
AU2014386718B2 (en) Use of benzoic acid salt in the manufactue of a composition for preventing or treating dementia or mild cognitive impairment
Bhandari et al. Resveratrol suppresses neuroinflammation in the experimental paradigm of autism spectrum disorders
Chu Alzheimer’s disease: early diagnosis and treatment
Moretti et al. Depression and Alzheimer's disease: symptom or comorbidity?
Ferreri et al. Motor cortex excitability in Alzheimer's disease: a transcranial magnetic stimulation follow-up study
JP6608858B2 (en) Use of L-carnitine, its salts and derivatives for reducing or preventing fatigue and for improving cognitive function
JP6550426B2 (en) Use of benzoate for producing a composition for preventing or treating dementia or mild cognitive impairment
Bhattacharya et al. Attention enhancing effects of methylphenidate are age-dependent
TWI573588B (en) Use of benzoic acid salt in the manufacture of a pharmaceutical composition for treating dementia or mild cognitive impairment
JP2008542263A (en) Cognitive ability improver
US20240016875A1 (en) Shan-zha for the treatment of depression and anxiety disorders
US20170216234A1 (en) Benzoates for use in treating dementia
Mintzer The search for better noncholinergic treatment options for Alzheimer's disease
Parikh et al. Current trends and updates in the treatment of Alzheimer's disease
Öhman Effect of exercise on cognition, physical functioning, fall rate, and neuropsychiatric symptoms in people with dementia
Haworth Gait, aging and dementia
Luijpen et al. Effects of transcutaneous electrical nerve stimulation (TENS) on memory in elderly with mild cognitive impairment
Ayati et al. Advances in treatment of mild cognitive impairment (MCI) and dementia: A review of promising non-pharmaceutical modalities
Devanand Depression in dementia
Alizadeh et al. The Effect of Pharmacotherapy Combined with Speech Therapy on Functional Recovery from Aphasia
BR102022006625A2 (en) FOOD SUPPLEMENT AND ITS USE
Lake et al. Integrative assessment and treatment of cognitive impairment
Gupta et al. Cognition Enhancers: A Review
Ljubenkov ENSURING SAFETY

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 2902498

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2016510654

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2014386718

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2120140000637

Country of ref document: DE

Ref document number: 212014000063

Country of ref document: DE

WWE Wipo information: entry into national phase

Ref document number: 2015/12039

Country of ref document: TR

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14887259

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 20167003184

Country of ref document: KR

Kind code of ref document: A

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205N DATED 13/02/2017)

122 Ep: pct application non-entry in european phase

Ref document number: 14887259

Country of ref document: EP

Kind code of ref document: A1