WO2022017923A1 - Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities - Google Patents

Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities Download PDF

Info

Publication number
WO2022017923A1
WO2022017923A1 PCT/EP2021/069829 EP2021069829W WO2022017923A1 WO 2022017923 A1 WO2022017923 A1 WO 2022017923A1 EP 2021069829 W EP2021069829 W EP 2021069829W WO 2022017923 A1 WO2022017923 A1 WO 2022017923A1
Authority
WO
WIPO (PCT)
Prior art keywords
cbd
preparation
seizures
thc
cannabinoids
Prior art date
Application number
PCT/EP2021/069829
Other languages
French (fr)
Inventor
Daniel Adam CHECKETTS
Kevin James CRAIG
Original Assignee
GW Research Limited
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 GW Research Limited filed Critical GW Research Limited
Publication of WO2022017923A1 publication Critical patent/WO2022017923A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/05Phenols
    • 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
    • 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
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4015Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil having oxo groups directly attached to the heterocyclic ring, e.g. piracetam, ethosuximide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • A61P25/12Antiepileptics; Anticonvulsants for grand-mal

Definitions

  • Focal (partial) seizures where the seizure originates within networks limited to only one hemisphere, are also split into sub-categories.
  • the seizure is characterized according to one or more features of the seizure, including aura, motor, autonomic and awareness / responsiveness.
  • a seizure begins as a localized seizure and rapidly evolves to be distributed within bilateral networks this seizure is known as a bilateral convulsive seizure, which is the proposed terminology to replace secondary generalized seizures (generalized seizures that have evolved from focal seizures and are no longer remain localized).
  • WO 2019/145700 relates to CBD for use in the reduction of seizures in treatment- resistant epilepsy where the CBD is administered in combination with an anti-epileptic drug that works via GABA receptor agonism such as stiripentol.
  • Phytocannabinoids can be obtained as either the neutral (decarboxylated form) or the carboxylic acid form depending on the method used to extract the cannabinoids. For example, it is known that heating the carboxylic acid form will cause most of the carboxylic acid form to decarboxylate into the neutral form.
  • the drug substance used is a liquid carbon dioxide extract of high-CBD containing chemotypes of Cannabis sativa L. which had been further purified by a solvent crystallization method to yield CBD.
  • the crystallisation process specifically removes other cannabinoids and plant components to yield greater than or equal to 95% CBD.
  • CBD is highly purified because it is produced from a cannabis plant rather than synthetically there is a small number of other cannabinoids which are co-produced and co-extracted with the CBD. Details of these cannabinoids and the quantities in which they are present in the medication are as described in Table A below.
  • the purity of the botanically derived purified CBD preparation was greater than or equal to 98%.
  • the botanically derived purified CBD includes THC and other cannabinoids, e.g., CBDA, CBDV, CBD-C1, and CBD-C4.
  • the CBD preparation comprises not more than 0.15% THC based on total amount of cannabinoid in the preparation.
  • the CBD preparation comprises about 0.01% to about 0.1% THC based on total amount of cannabinoid in the preparation.
  • the CBD preparation comprises about 0.02% to about 0.05% THC based on total amount of cannabinoid in the preparation.
  • the CBD preparation comprises about 0.2% to about 1.0%
  • the CBD preparation comprises about 0.3% to about 0.5% CBD-C4 based on total amount of cannabinoid in the preparation. In some embodiments, the CBD preparation comprises about 0.3% to about 0.4% CBD-C4 based on total amount of cannabinoid in the preparation. [0056] In some embodiments, the CBD preparation comprises about 0.1% to about 0.15% CBD-C1 based on total amount of cannabinoid in the preparation.
  • High CBD chemovars were grown, harvested, dried, baled and stored in a dry room until required.
  • the botanical raw material (BRM) was finely chopped using an Apex mill fitted with a 1 mm screen. The milled BRM was stored in a freezer prior to extraction.
  • the manufacturing steps to produce the botanically derived purified CBD preparation from BDS were as follows: a) Crystallization using C 5 -C 12 straight chain or branched alkane b) Filtration c) Vacuum drying [0063]
  • the BDS produced using the methodology above was dispersed in C5-C12 straight chain or branched alkane. The mixture was manually agitated to break up any lumps and the sealed container then placed in a freezer for approximately 48 hours. The crystals were isolated via vacuum filtration, washed with aliquots of cold C5-C12 straight chain or branched alkane, and dried under a vacuum of ⁇ 10mb at a temperature of 60°C until dry.
  • the botanically derived purified CBD preparation was stored in a freezer at -20°C in a pharmaceutical grade stainless steel container, with FDA food grade approved silicone seal and clamps.
  • the botanically derived purified CBD used in the clinical trial described in the invention comprises greater than or equal to 98% (w/w) CBD and less than or equal to 2% (w/w) of other cannabinoids.
  • the other cannabinoids present are THC at a concentration of less than or equal to 0.1% (w/w); CBD-C1 at a concentration of less than or equal to 0.15% (w/w); CBDV at a concentration of less than or equal to 0.8% (w/w); and CBD-C4 at a concentration of less than or equal to 0.4% (w/w).
  • this study signifies the use of CBD for treatment of seizures associated with cardiofaciocutaneous syndrome.
  • Seizure types include tonic-clonic and myoclonic seizures which were decreased by significant rates up to 100%.

Abstract

The present invention relates to the use of cannabidiol (CBD) for the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities. In particular the seizures associated with rare epilepsy syndromes that are treated are those which are experienced in patients diagnosed with cardiofaciocutaneous syndrome. In a further embodiment the types of seizures include tonic-clonic and myoclonic. Preferably the dose of CBD is between 5 mg/kg/day to 50 mg/kg/day.

Description

USE OF CANNABIDIOL IN THE TREATMENT OF SEIZURES ASSOCIATED WITH RARE EPILEPSY SYNDROMES RELATED TO GENETIC ABNORMALITIES
FIELD OF THE INVENTION
[0001] The present invention relates to the use of cannabidiol (CBD) for the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities. In particular the seizures associated with rare epilepsy syndromes that are treated are those which are experienced in patients diagnosed with cardiofaciocutaneous syndrome. In a further embodiment the types of seizures include tonic-clonic and myoclonic. Preferably the dose of CBD is between 5 mg/kg/day to 50 mg/kg/day.
[0002] In a further embodiment the CBD used is in the form of a highly purified extract of cannabis such that the CBD is present at greater than 95% of the total extract (w/w) and the cannabinoid tetrahydrocannabinol (THC) has been substantially removed, to a level of not more than 0.15% (w/w).
[0003] Preferably the CBD used is in the form of a botanically derived purified CBD which comprises greater than or equal to 98% (w/w) CBD and less than or equal to 2% (w/w) of other cannabinoids. More preferably the other cannabinoids present are THC at a concentration of less than or equal to 0.1% (w/w); CBD-C1 at a concentration of less than or equal to 0.15% (w/w); CBDV at a concentration of less than or equal to 0.8% (w/w); and CBD-C4 at a concentration of less than or equal to 0.4% (w/w). The botanically derived purified CBD preferably also comprises a mixture of both trans-THC and cis-THC. Alternatively, a synthetically produced CBD is used.
[0004] Most preferably the other cannabinoids present are THC at a concentration of about 0.01% to about 0.1% (w/w); CBD-C1 at a concentration of about 0.1% to about 0.15% (w/w); CBDV at a concentration of about 0.2% to about 0.8% (w/w); and CBD-C4 at a concentration of about 0.3% to about 0.4% (w/w). Most preferably still the THC is present at a concentration of about 0.02% to about 0.05% (w/w).
[0005] Where the CBD is given concomitantly with one or more other anti-epileptic drugs (AED), the CBD may be formulated for administration separately, sequentially or simultaneously with one or more AED or the combination may be provided in a single dosage form.
BACKGROUND TO THE INVENTION
[0006] Epilepsy occurs in approximately 1% of the population worldwide, (Thurman et al., 2011) of which 70% are able to adequately control their symptoms with the available existing anti-epileptic drugs (AED). However, 30% of this patient group, (Eadie etal., 2012), are unable to obtain seizure freedom from the AED that are available and as such are termed as suffering from intractable or “treatment-resistant epilepsy” (TRE).
[0007] Intractable or treatment-resistant epilepsy was defined in 2009 by the International League Against Epilepsy (I LAE) as “failure of adequate trials of two tolerated and appropriately chosen and used AED schedules (whether as monotherapies or in combination) to achieve sustained seizure freedom ” (Kwan et al., 2009).
[0008] Individuals who develop epilepsy during the first few years of life are often difficult to treat and as such are often termed treatment resistant. Children who undergo frequent seizures in childhood are often left with neurological damage which can cause cognitive, behavioral and motor delays.
[0009] Childhood epilepsy is a relatively common neurological disorder in children and young adults with a prevalence of approximately 700 per 100,000. This is twice the number of epileptic adults per population.
[0010] When a child or young adult presents with a seizure, investigations are normally undertaken in order to investigate the cause. Childhood epilepsy can be caused by many different syndromes and genetic mutations and as such diagnosis for these children may take some time.
[0011] The main symptom of epilepsy is repeated seizures. In order to determine the type of epilepsy or the epileptic syndrome that a patient is suffering from an investigation into the type of seizures that the patient is experiencing is undertaken. Clinical observations and electroencephalography (EEG) tests are conducted and the type(s) of seizures are classified according to the ILEA classification.
[0012] Generalized seizures, where the seizure arises within and rapidly engages bilaterally distributed networks, can be split into six subtypes: tonic-clonic (grand mal) seizures; absence (petit mal) seizures; clonic seizures; tonic seizures; atonic seizures and myoclonic seizures.
[0013] Focal (partial) seizures where the seizure originates within networks limited to only one hemisphere, are also split into sub-categories. Here the seizure is characterized according to one or more features of the seizure, including aura, motor, autonomic and awareness / responsiveness. Where a seizure begins as a localized seizure and rapidly evolves to be distributed within bilateral networks this seizure is known as a bilateral convulsive seizure, which is the proposed terminology to replace secondary generalized seizures (generalized seizures that have evolved from focal seizures and are no longer remain localized).
[0014] Focal seizures where the subject’s awareness / responsiveness is altered are referred to as focal seizures with impairment and focal seizures where the awareness or responsiveness of the subject is not impaired are referred to as focal seizures without impairment. [0015] Cardiofaciocutaneous syndrome is a disorder that affects many parts of the body, particularly the heart, facial features, and the skin and hair. People with this condition also have delayed development and intellectual disability, usually ranging from moderate to severe.
[0016] Heart defects occur in most people with cardiofaciocutaneous syndrome. The heart problems most commonly associated with this condition include malformations of one of the heart valves that impairs blood flow from the heart to the lungs (pulmonic stenosis), a hole between the two upper chambers of the heart (atrial septal defect), and a form of heart disease that enlarges and weakens the heart muscle (hypertrophic cardiomyopathy).
[0017] Cardiofaciocutaneous syndrome is also characterized by distinctive facial features. These include a high forehead that narrows at the temples, a short nose, widely spaced eyes (ocular hypertelorism), outside corners of the eyes that point downward (down-slanting palpebral fissures), droopy eyelids (ptosis), a small chin, and low-set ears. Overall, the face is broad and long, and the facial features are sometimes described as "coarse."
[0018] Skin abnormalities occur in almost everyone with cardiofaciocutaneous syndrome. Many affected people have dry, rough skin; dark-coloured moles (nevi); wrinkled palms and soles; and a skin condition called keratosis pilaris, which causes small bumps to form on the arms, legs, and face. People with cardiofaciocutaneous syndrome also tend to have thin, dry, curly hair and sparse or absent eyelashes and eyebrows.
[0019] Infants with cardiofaciocutaneous syndrome typically have weak muscle tone (hypotonia), feeding difficulties, and a failure to grow and gain weight at the normal rate (failure to thrive). Additional features of this disorder in children and adults can include an unusually large head (macrocephaly), short stature, problems with vision, and seizures.
[0020] Cardiofaciocutaneous syndrome can be caused by mutations in several genes. Mutations in the BRAF gene are most common, accounting for 75 to 80 percent of all cases. Another 10 to 15 percent of cases result from mutations in one of two similar genes, MAP2K1 and MAP2K2. Fewer than 5 percent of cases are caused by mutations in the KRAS gene. [0021] Cannabidiol (CBD), a non-psychoactive derivative from the cannabis plant, has demonstrated anti-convulsant properties in several anecdotal reports, pre-clinical and clinical studies both in animal models and humans. Three randomized control trials showed efficacy of the purified pharmaceutical formulation of CBD in patients with Dravet and Lennox-Gastaut syndrome.
[0022] Based on these three trials, a botanically derived purified CBD preparation was approved by FDA in June 2018 for the treatment of seizures associated with Dravet and Lennox-Gastaut syndromes.
[0023] A news article in 2016 discussed the use of cannabis oil in a child suffering from cardiofaciocutaneous syndrome.1 It is disclosed that there was no THC contained in the oil. [0024] WO 2019/145700 relates to CBD for use in the reduction of seizures in treatment- resistant epilepsy where the CBD is administered in combination with an anti-epileptic drug that works via GABA receptor agonism such as stiripentol.
[0025] The applicant has found by way of an open label, expanded-access program that treatment with CBD resulted in a significant reduction in atonic and absence seizures in a patient with auriculotemporal syndrome.
BRIEF SUMMARY OF THE DISCLOSURE
[0026] In accordance with a first aspect of the present invention there is provided a cannabidiol (CBD) preparation for use in the treatment of cardiofaciocutaneous syndrome.
[0027] In a further embodiment, the seizures associated with cardiofaciocutaneous syndrome are tonic-clonic and myoclonic seizures.
[0028] In a further embodiment, the CBD preparation comprises greater than 95% (w/w) CBD and not more than 0.15% (w/w) tetrahydrocannabinol (THC).
[0029] Preferably the CBD preparation comprises greater than or equal to 98% (w/w) CBD and less than or equal to 2% (w/w) other cannabinoids, wherein the less than or equal to 2% (w/w) other cannabinoids comprise the cannabinoids tetrahydrocannabinol (THC); cannabidiol- C1 (CBD-C1); cannabidivarin (CBDV); and cannabidiol-C4 (CBD-C4), and wherein the THC is present as a mixture of trans-THC and cis-THC.
[0030] Preferably the CBD preparation is used in combination with one or more concomitant anti-epileptic drugs (AED).
[0031] Preferably the one or more AED is selected from the group consisting of: levetiracetam, pregabalin and lacosamide.
[0032] In one embodiment the CBD is present is isolated from cannabis plant material. Preferably at least a portion of at least one of the cannabinoids present in the CBD preparation is isolated from cannabis plant material.
[0033] In a further embodiment the CBD is present as a synthetic preparation. Preferably at least a portion of at least one of the cannabinoids present in the CBD preparation is prepared synthetically.
[0034] Preferably the dose of CBD is greater than 5 mg/kg/day. More preferably the dose of CBD is 20 mg/kg/day. More preferably the dose of CBD is 25 mg/kg/day. More preferably the dose of CBD is 50 mg/kg/day. [0035] In accordance with a second aspect of the present invention there is provided a method of treating seizures associated with cardiofaciocutaneous syndrome comprising administering a cannabidiol (CBD) preparation to the subject in need thereof.
DEFINITIONS
[0036] Definitions of some of the terms used to describe the invention are detailed below:
[0037] Over 100 different cannabinoids have been identified, see for example, Handbook of Cannabis, Roger Pertwee, Chapter 1, pages 3 to 15. These cannabinoids can be split into different groups as follows: Phytocannabinoids; Endocannabinoids and Synthetic cannabinoids (which may be novel cannabinoids or synthetically produced phytocannabinoids or endocannabinoids).
[0038] “Phytocannabinoids” are cannabinoids that originate from nature and can be found in the cannabis plant. The phytocannabinoids can be isolated from plants to produce a highly purified extract or can be reproduced synthetically.
[0039] “Highly purified cannabinoids” are defined as cannabinoids that have been extracted from the cannabis plant and purified to the extent that other cannabinoids and non-cannabinoid components that are co-extracted with the cannabinoids have been removed, such that the highly purified cannabinoid is greater than or equal to 95% (w/w) pure.
[0040] “Synthetic cannabinoids” are compounds that have a cannabinoid or cannabinoid-like structure and are manufactured using chemical means rather than by the plant.
[0041] Phytocannabinoids can be obtained as either the neutral (decarboxylated form) or the carboxylic acid form depending on the method used to extract the cannabinoids. For example, it is known that heating the carboxylic acid form will cause most of the carboxylic acid form to decarboxylate into the neutral form.
[0042] “Treatment-resistant epilepsy” (TRE) or “intractable epilepsy” is defined as per the I LAE guidance of 2009 as epilepsy that is not adequately controlled by trials of one or more AED.
[0043] “Tonic-clonic seizures” consist of two phases: the tonic phase and the clonic phase. In the tonic phase the body becomes entire rigid, and in the clonic phase there is uncontrolled jerking. Tonic-clonic seizures may or may not be preceded by an aura, and are often followed by headache, confusion, and sleep. They may last mere seconds or continue for several minutes. These seizures are also known as a grand mal seizure.
[0044] “Myoclonic seizures” are characterised by a ‘muscle jerk’. Myoclonic seizures are brief but can happen in clusters (many happening close together in time) and often happen shortly after waking. In myoclonic seizures the person is conscious, but they are classified as generalised seizures.
DETAILED DESCRIPTION
PREPARATION OF HIGHLY PURIFIED CBD EXTRACT
[0045] The following describes the production of the highly-purified (>95% w/w) cannabidiol extract which has a known and constant composition.
[0046] In summary the drug substance used is a liquid carbon dioxide extract of high-CBD containing chemotypes of Cannabis sativa L. which had been further purified by a solvent crystallization method to yield CBD. The crystallisation process specifically removes other cannabinoids and plant components to yield greater than or equal to 95% CBD. Although the CBD is highly purified because it is produced from a cannabis plant rather than synthetically there is a small number of other cannabinoids which are co-produced and co-extracted with the CBD. Details of these cannabinoids and the quantities in which they are present in the medication are as described in Table A below.
Table A: Composition of highly purified CBD extract
Figure imgf000007_0001
> - greater than NMT - not more than
PREPARATION OF BOTANICALLY DERIVED PURIFIED CBD
[0047] The following describes the production of the botanically derived purified CBD which comprises greater than or equal to 98% w/w CBD and less than or equal to other cannabinoids was used in the open label, expanded-access program described in Example 1 below.
[0048] In summary the drug substance used in the trials is a liquid carbon dioxide extract of high-CBD containing chemotypes of Cannabis sativa L. which had been further purified by a solvent crystallization method to yield CBD. The crystallisation process specifically removes other cannabinoids and plant components to yield greater than 95% CBD w/w, typically greater than 98% w/w. [0049] The Cannabis sativa L. plants are grown, harvested, and processed to produce a botanical extract (intermediate) and then purified by crystallization to yield the CBD (botanically derived purified CBD).
[0050] The plant starting material is referred to as Botanical Raw Material (BRM); the botanical extract is the intermediate; and the active pharmaceutical ingredient (API) is CBD, the drug substance.
[0051] All parts of the process are controlled by specifications. The botanical raw material specification is described in Table B and the CBD API is described in Table C. Table B: CBD botanical raw material specification
Figure imgf000008_0001
Table C: Specification of an exemplary botanically derived purified CBD preparation
Figure imgf000009_0001
[0052] The purity of the botanically derived purified CBD preparation was greater than or equal to 98%. The botanically derived purified CBD includes THC and other cannabinoids, e.g., CBDA, CBDV, CBD-C1, and CBD-C4. [0053] In some embodiments, the CBD preparation comprises not more than 0.15% THC based on total amount of cannabinoid in the preparation. In some embodiments, the CBD preparation comprises about 0.01% to about 0.1% THC based on total amount of cannabinoid in the preparation. In some embodiments, the CBD preparation comprises about 0.02% to about 0.05% THC based on total amount of cannabinoid in the preparation. [0054] In some embodiments, the CBD preparation comprises about 0.2% to about 1.0%
CBDV based on total amount of cannabinoid in the preparation. In some embodiments, the CBD preparation comprises about 0.2% to about 0.8% CBDV based on total amount of cannabinoid in the preparation.
[0055] In some embodiments, the CBD preparation comprises about 0.3% to about 0.5% CBD-C4 based on total amount of cannabinoid in the preparation. In some embodiments, the CBD preparation comprises about 0.3% to about 0.4% CBD-C4 based on total amount of cannabinoid in the preparation. [0056] In some embodiments, the CBD preparation comprises about 0.1% to about 0.15% CBD-C1 based on total amount of cannabinoid in the preparation.
[0057] Distinct chemotypes of the Cannabis sativa L. plant have been produced to maximize the output of the specific chemical constituents, the cannabinoids. Certain chemovars produce predominantly CBD. Only the (-)-trans isomer of CBD is believed to occur naturally. During purification, the stereochemistry of CBD is not affected.
Production of CBD botanical drug substance
[0058] An overview of the steps to produce a botanical extract, the intermediate, are as follows: a) Growing b) Direct drying c) Decarboxylation d) Extraction - using liquid CO2 e) Winterization using ethanol f) Filtration g) Evaporation
[0059] High CBD chemovars were grown, harvested, dried, baled and stored in a dry room until required. The botanical raw material (BRM) was finely chopped using an Apex mill fitted with a 1 mm screen. The milled BRM was stored in a freezer prior to extraction.
[0060] Decarboxylation of CBDA to CBD was carried out using heat. BRM was decarboxylated at 115°C for 60 minutes.
[0061] Extraction was performed using liquid CO2 to produce botanical drug substance (BDS), which was then crystalized to produce the test material. The crude CBD BDS was winterized to refine the extract under standard conditions (2 volumes of ethanol at -20°C for approximately 50 hours). The precipitated waxes were removed by filtration and the solvent was removed to yield the BDS.
Production of botanically derived purified CBD preparation
[0062] The manufacturing steps to produce the botanically derived purified CBD preparation from BDS were as follows: a) Crystallization using C5-C12 straight chain or branched alkane b) Filtration c) Vacuum drying [0063] The BDS produced using the methodology above was dispersed in C5-C12 straight chain or branched alkane. The mixture was manually agitated to break up any lumps and the sealed container then placed in a freezer for approximately 48 hours. The crystals were isolated via vacuum filtration, washed with aliquots of cold C5-C12 straight chain or branched alkane, and dried under a vacuum of <10mb at a temperature of 60°C until dry. The botanically derived purified CBD preparation was stored in a freezer at -20°C in a pharmaceutical grade stainless steel container, with FDA food grade approved silicone seal and clamps.
Physicochemical properties of the botanically derived purified CBD [0064] The botanically derived purified CBD used in the clinical trial described in the invention comprises greater than or equal to 98% (w/w) CBD and less than or equal to 2% (w/w) of other cannabinoids. The other cannabinoids present are THC at a concentration of less than or equal to 0.1% (w/w); CBD-C1 at a concentration of less than or equal to 0.15% (w/w); CBDV at a concentration of less than or equal to 0.8% (w/w); and CBD-C4 at a concentration of less than or equal to 0.4% (w/w).
[0065] The botanically derived purified CBD used additionally comprises a mixture of both trans-THC and cis-THC. It was found that the ratio of the trans-THC to cis-THC is altered and can be controlled by the processing and purification process, ranging from 3.3:1 (trans-THC:cis- THC) in its unrefined decarboxylated state to 0.8:1 (trans-THC:cis-THC) when highly purified. [0066] Furthermore, the cis-THC found in botanically derived purified CBD is present as a mixture of both the (+)-cis-THC and the (-)-cis-THC isoforms.
[0067] Clearly a CBD preparation could be produced synthetically by producing a composition with duplicate components.
[0068] Example 1 below describes the use of a botanically derived purified CBD in an open label, expanded-access program to investigate the clinical efficacy and safety of purified pharmaceutical cannabidiol formulation (CBD) in the treatment of cardiofaciocutaneous syndrome.
EXAMPLE 1: CLINICAL EFFICACY AND SAFETY OF PURIFIED PHARMACEUTICAL CANNABIDIOL (CBD) IN THE TREATMENT OF A PATIENT DIAGNOSED WITH CARDIOFACIOCUTANEOUS SYNDROME
Study design [0069] The subject was required to be on one or more AEDs at stable doses for a minimum of two weeks prior to baseline and to have stable vagus nerve stimulation (VNS) settings and ketogenic diet ratios for a minimum of four weeks prior to baseline.
[0070] The patient was administered botanically derived purified CBD in a 100 mg/ml_ sesame oil-based solution at an initial dose of 5 milligrams per kilogram per day (mg/kg/day) in two divided doses. Dose was then increased weekly by 5mg/kg/day to a goal of 20 to 25 mg/kg/day.
[0071] A maximum dose of 50 mg/kg/day could be utilised should the patient require it to achieve seizure control; further weekly titration by 5mg/kg/day is additionally required for these higher doses.
[0072] There was one patient in this study, and they received CBD for 144 weeks. Modifications were made to concomitant AEDs as per clinical indication.
[0073] Seizure frequency, intensity, and duration were recorded by caregivers in a diary during a baseline period of at least 28 days. Changes in seizure frequency relative to baseline were calculated after at least 2 weeks and at defined timepoints of treatment.
Statistical Methods:
[0074] Patients may be defined as responders if they had more than 50% reduction in seizure frequency compared to baseline. The percent change in seizure frequency was calculated as follows:
% change= ((weekly seizure frequency time interval)-( weekly seizure frequency Baseline)) x100 seizure (weekly seizure frequency Baseline) frequency
[0075] The percent change of seizure frequency may be calculated for any time interval where seizure number has been recorded. For the purpose of this example the percent change of seizure frequency for the end of the treatment period was calculated as follows:
% reduction = ((weekly seizure frequency Baseline) - (weekly seizure frequency End)) x100 seizure frequency (weekly seizure frequency Baseline)
Results
Patient description [0076] There was one patient enrolled in the open label, expanded-access program which was diagnosed with cardiofaciocutaneous syndrome. This patient experienced tonic-clonic and myoclonic seizures and was taking several concomitant AEDs.
[0077] The age of the patient was 13 years old, and she was female as detailed in Table 1 below.
Table 1: Patient demographics, seizure type and concomitant medication
Figure imgf000013_0001
LEV = levetiracetam, PGB = pregabalin, LCS = lacosamide
Study medication and concomitant medications
[0078] The patient on the study was titrated up to a dose of 25mg/kg/day of CBD. The patient was on three concomitant AEDs at the time of starting CBD. Clinical changes
[0079] Table 2 illustrates the seizure frequency for the patient as well as the dose of CBD given.
Table 2: Seizure frequency data for Patient 1
Figure imgf000013_0002
Figure imgf000014_0001
[0080] Patient 1 was treated for 144 weeks and experienced a 100% reduction in myoclonic seizures over the treatment period. [0081] Overall, CBD was particularly effective in reducing the frequency myoclonic seizures. The patient became completely free of myoclonic seizures after 36 weeks of treatment.
Conclusions [0082] These data indicate that CBD was able to significantly reduce the number of seizures associated with cardiofaciocutaneous syndrome. Clearly the treatment is of significant benefit in this rare syndrome given the high response rate experienced this patient.
[0083] In conclusion, this study signifies the use of CBD for treatment of seizures associated with cardiofaciocutaneous syndrome. Seizure types include tonic-clonic and myoclonic seizures which were decreased by significant rates up to 100%.
REFERENCES
1. Draaisma (2016) "Nurses won't give cannabis oil to son who suffers seizures, Toronto mom says." CBC News https://www.cbc.ca/news/canada/toronto/cannabis- oilnurses-ontario-sickkids-seizures-1.3739949

Claims

1. A cannabidiol (CBD) preparation for use in the treatment of seizures associated with cardiofaciocutaneous syndrome, wherein the CBD preparation comprises greater than 95% (w/w) CBD and between 0.01 and 0.15% (w/w) tetrahydrocannabinol (THC).
2. A CBD preparation for use according to claim 1, wherein the seizures associated with cardiofaciocutaneous syndrome are tonic-clonic and myoclonic seizures.
3. A CBD preparation for use according to any of the preceding claims, wherein the CBD preparation comprises greater than or equal to 98% (w/w) CBD and less than or equal to 2% (w/w) other cannabinoids, wherein the less than or equal to 2% (w/w) other cannabinoids comprise the cannabinoids tetrahydrocannabinol (THC); cannabidiol-C1 (CBD-C1); cannabidivarin (CBDV); and cannabidiol-C4 (CBD-C4), and wherein the THC is present as a mixture of trans-THC and cis-THC.
4. A CBD preparation to any of the preceding claims, wherein the CBD preparation is used in combination with one or more concomitant anti-epileptic drugs (AED).
5. A CBD preparation for use according to claim 4, wherein the one or more AED is selected from a group consisting of: levetiracetam, pregabalin and lacosamide.
6. A CBD preparation for use according to any of the preceding claims, wherein the CBD is present is isolated from cannabis plant material.
7. A CBD preparation for use according to any of the preceding claims, wherein at least a portion of at least one of the cannabinoids present in the CBD preparation is isolated from cannabis plant material.
8. A CBD preparation for use according to claims 1 to 5, wherein the CBD is present as a synthetic preparation.
9. A CBD preparation for use according to claim 8, wherein at least a portion of at least one of the cannabinoids present in the CBD preparation is prepared synthetically.
10. A CBD preparation for use according to any of the preceding claims, wherein the dose of CBD is greater than 5 mg/kg/day.
11. A CBD preparation for use according to any of the preceding claims, wherein the dose of CBD is 20 mg/kg/day.
12. A CBD preparation for use according to any of the preceding claims, wherein the dose of CBD is 25 mg/kg/day.
13. A CBD preparation for use according to any of the preceding claims, wherein the dose of
CBD is 50 mg/kg/day.
A method of treating seizures associated with cardiofaciocutaneous syndrome comprising administering a cannabidiol (CBD) preparation to the subject in need thereof.
PCT/EP2021/069829 2020-07-20 2021-07-15 Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities WO2022017923A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB2011124.1A GB2597282A (en) 2020-07-20 2020-07-20 Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities
GB2011124.1 2020-07-20

Publications (1)

Publication Number Publication Date
WO2022017923A1 true WO2022017923A1 (en) 2022-01-27

Family

ID=72339059

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2021/069829 WO2022017923A1 (en) 2020-07-20 2021-07-15 Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities

Country Status (2)

Country Link
GB (1) GB2597282A (en)
WO (1) WO2022017923A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015193668A1 (en) * 2014-06-17 2015-12-23 Gw Pharma Limited Use of cannabidiol in the treatment of epilepsy
GB2531280A (en) * 2014-10-14 2016-04-20 Gw Pharma Ltd Use of cannabidiol in the treatment of intractable epilepsy
WO2019145700A1 (en) 2018-01-24 2019-08-01 GW Research Limited Use of cannabinoids in the treatment of epilepsy
WO2019207319A1 (en) * 2018-04-27 2019-10-31 GW Research Limited Cannabidiol preparations and its uses

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015193668A1 (en) * 2014-06-17 2015-12-23 Gw Pharma Limited Use of cannabidiol in the treatment of epilepsy
GB2531280A (en) * 2014-10-14 2016-04-20 Gw Pharma Ltd Use of cannabidiol in the treatment of intractable epilepsy
WO2019145700A1 (en) 2018-01-24 2019-08-01 GW Research Limited Use of cannabinoids in the treatment of epilepsy
WO2019207319A1 (en) * 2018-04-27 2019-10-31 GW Research Limited Cannabidiol preparations and its uses

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DRAAISMA MURIEL: "Nurses won't give cannabis oil to son who suffers seizures, Toronto mom says | CBC News", 29 August 2016 (2016-08-29), XP055835468, Retrieved from the Internet <URL:https://www.cbc.ca/news/canada/toronto/cannabis-oil-nurses-ontario-sickkids-seizures-1.3739949> [retrieved on 20210827] *

Also Published As

Publication number Publication date
GB202011124D0 (en) 2020-09-02
GB2597282A (en) 2022-01-26

Similar Documents

Publication Publication Date Title
EP4182024A1 (en) Use of cannabidiol in the treatment of seizures associated with mutations in the syngap1 gene
WO2022017923A1 (en) Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities
WO2022017947A1 (en) Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities
WO2022017930A1 (en) Use of cannabidiol in the treatment of seizures associated with jeavon&#39;s syndrome
WO2022017938A1 (en) Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities
EP4181893A1 (en) Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities
EP4181888A1 (en) Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to structural abnormalities of the brain
WO2022017954A1 (en) Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to structural abnormalities of the brain
EP4181902A1 (en) Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities
WO2022017955A1 (en) Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to structural abnormalities of the brain
WO2022017927A1 (en) Use of cannabidiol in the treatment of seizures associated with perisylvian fissure syndrome
WO2022017936A1 (en) Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to structural abnormalities of the brain
WO2022017931A1 (en) Use of cannabidiol in the treatment of seizures associated with ohtahara syndrome or ohtahara syndrome secondary to aimp1 mutation
EP4181905A1 (en) Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities
WO2022017948A1 (en) Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities
WO2022017929A1 (en) Use of cannabidiol in the treatment of seizures associated lissencephaly
WO2022017925A1 (en) Use of cannabidiol in the treatment of seizures associated with multifocal epilepsy syndrome
EP4181897A1 (en) Use of cannabidiol in the treatment of seizures associated with chrna4 mutation
WO2022017944A1 (en) Use of cannabidiol in the treatment of seizures associated with bilateral cerebral dysgenesis
WO2022017950A1 (en) Use of cannabidiol in the treatment of seizures associated with bilateral mesial temporal sclerosis
WO2022017958A1 (en) Use of cannabidiol in the treatment of seizures associated with shaken baby syndrome
WO2022017953A1 (en) Use of cannabidiol in the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities
EP4181896A1 (en) Cannabidiol for use in the treatment of seizures associated with rare epilepsy syndromes related to genetic abnormalities
WO2022017951A1 (en) Use of cannabidiol in the treatment of seizures associated cask-related disorders
WO2022017943A1 (en) Use of cannabidiol in the treatment seizures associated with arginase deficiency

Legal Events

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

Ref document number: 21749781

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21749781

Country of ref document: EP

Kind code of ref document: A1