USE OF CANNABINOIDS IN THE TREATMENT OF EPILEPSY

Information

  • Patent Application
  • 20220249396
  • Publication Number
    20220249396
  • Date Filed
    December 16, 2021
    2 years ago
  • Date Published
    August 11, 2022
    a year ago
Abstract
The present invention relates to the use of cannabidiol (CBD) in the treatment of patients with childhood-onset epilepsy who are concurrently taking one or more antiepileptic drugs that works via GABA receptor agonism. Preferably the AED is stiripentol. Preferably the CBD used is in the form of a highly purified extract of cannabis such that the CBD is present at greater than 98% of the total extract (w/w) and the other components of the extract are characterized. In particular the cannabinoid tetrahydrocannabinol (THC) has been substantially removed, to a level of not more than 0.15% (w/w) and the propyl analogue of CBD, cannabidivarin, (CBDV) is present in amounts of up to 1%. Alternatively, the CBD may be a synthetically produced CBD.
Description
FIELD OF THE INVENTION

The present invention relates to the use of cannabidiol (CBD) in the treatment of patients with childhood-onset epilepsy who are concurrently taking one or more antiepileptic drugs that works via GABA receptor agonism. Preferably the AED is stiripentol.


Preferably the CBD used is in the form of a highly purified extract of cannabis such that the CBD is present at greater than 98% of the total extract (w/w) and the other components of the extract are characterised. In particular the cannabinoid tetrahydrocannabinol (THC) has been substantially removed, to a level of not more than 0.15% (w/w) and the propyl analogue of CBD, cannabidivarin, (CBDV) is present in amounts of up to 1%. Alternatively, the CBD may be a synthetically produced CBD.


BACKGROUND TO THE INVENTION

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 (AEDs). However, 30% of this patient group, (Eadie et at, 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).


Intractable or treatment-resistant epilepsy was defined in 2009 by the International League Against Epilepsy (ILAE) 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).


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.


Childhood-onset 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.


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.


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 ILAE classification described below.


The International classification of seizure types proposed by the ILAE was adopted in 1981 and a revised proposal was published by the ILAE in 2010 and has not yet superseded the 1981 classification. The 2010 proposal for revised terminology includes the proposed changes to replace the terminology of partial with focal. In addition, the term “simple partial seizure” has been replaced by the term “focal seizure where awareness/responsiveness is not impaired” and the term “complex partial seizure” has been replaced by the term “focal seizure where awareness/consciousness is impaired”.


Generalised 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.


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 Generalised Seizures (generalized seizures that have evolved from focal seizures and are no longer remain localized).


Epileptic syndromes often present with many different types of seizure and identifying the types of seizure that a patient is suffering from is important as many of the standard AEDs are targeted to treat or are only effective against a given seizure type/sub-type.


One such childhood epilepsy syndrome is Lennox-Gastaut syndrome (LGS). LGS is a severe form of epilepsy, where seizures usually begin before the age of 4. Seizure types, which vary among patients, include tonic (stiffening of the body, upward deviation of the eyes, dilation of the pupils, and altered respiratory patterns), atonic (brief loss of muscle tone and consciousness, causing abrupt falls), atypical absence (staring spells), and myoclonic (sudden muscle jerks). There may be periods of frequent seizures mixed with brief, relatively seizure-free periods.


Seizures in LGS are often described as “drop seizures”. Such drop seizures are defined as an attack or spell (atonic, tonic or tonic-clonic) involving the entire body, trunk or head that led or could have led to a fall, injury, slumping in a chair or hitting the patient's head on a surface.


Most patients with LGS experience some degree of impaired intellectual functioning or information processing, along with developmental delays, and behavioural disturbances.


LGS can be caused by brain malformations, perinatal asphyxia, severe head injury, central nervous system infection and inherited degenerative or metabolic conditions. In 30-35% of cases, no cause can be found.


The first line treatment for drop seizures, including the treatment of drop seizures in patients with LGS, usually comprises a broad-spectrum AED, such as sodium valproate often in combination with rufinamide or lamotrigine. Other AEDs that may be considered include felbamate, clobazam and topiramate.


Another childhood epilepsy syndrome is Dravet syndrome. Onset of Dravet syndrome almost always occurs during the first year of life with clonic and tonic-clonic seizures in previously healthy and developmentally normal infants (Dravet, 2011). Symptoms peak at about five months of age. Other seizures develop between one and four years of age such as prolonged focal dyscognitive seizures and brief absence seizures.


In diagnosing Dravet syndrome both focal and generalised seizures are considered to be mandatory, Dravet patients may also experience atypical absence seizures, myoclonic absence seizures, atonic seizures and non-convulsive status epilepticus.


Seizures progress to be frequent and treatment-resistant, meaning that the seizures do not respond well to treatment. They also tend to be prolonged, lasting more than 5 minutes. Prolonged seizures may lead to status epilepticus, which is a seizure that lasts more than 30 minutes, or seizures that occur in clusters, one after another.


Prognosis is poor and approximately 14% of children die during a seizure, because of infection, or suddenly due to uncertain causes, often because of the relentless neurological decline. Patients develop intellectual disability and life-long ongoing seizures. Intellectual impairment varies from severe in 50% patients, to moderate and mild intellectual disability each accounting for 25% of cases.


There are currently no FDA approved treatments specifically indicated for Dravet syndrome. The standard of care usually involves a combination of the following anticonvulsants: clobazam, clonazepam, levetiracetam, topiramate and valproic acid.


Stiripentol is approved in Europe for the treatment of Dravet syndrome in conjunction with clobazam and valproic acid. In the US, stiripentol was granted an Orphan Designation for the treatment of Dravet syndrome in 2008; however, the drug is not FDA approved.


Potent sodium channel blockers used to treat epilepsy actually increase seizure frequency in patients with Dravet Syndrome. The most common are phenytoin, carbamazepine, lamotrigine and rufinamide.


Management may also include a ketogenic diet, and physical and vagus nerve stimulation. In addition to anti-convulsive drugs, many patients with Dravet syndrome are treated with anti-psychotic drugs, stimulants, and drugs to treat insomnia.


Common AEDs defined by their mechanisms of action are described in the following tables:









TABLE 1







Examples of narrow spectrum AEDs









Narrow-spectrum




AED
Mechanism
Indication





Phenytoin
Sodium channel
Complex partial




Tonic-clonic


Phenobarbital
GABA/Calcium channel
Partial seizures




Tonic-clonic


Carbamazepine
Sodium channel
Partial seizures




Tonic-clonic




Mixed seizures


Oxcarbazepine
Sodium channel
Partial seizures




Tonic-clonic




Mixed seizures


Gabapentin
Calcium channel
Partial seizures




Mixed seizures


Pregabalin
Calcium channel
Adjunct therapy




for partial seizures




with or without




secondary




generalisation


Lacosamide
Sodium channel
Adjunct therapy




for partial seizures


Vigabatrin
GABA receptor
Secondarily generalized



agonism
tonic-clonic seizures




Partial seizures




Infantile spasms due




to West syndrome
















TABLE 2







Examples of broad spectrum AEDs











Broad-spectrum





AED
Mechanism
Indication






Valproic acid
GABA/Sodium
First-line treatment




channel
for tonic-clonic





seizures, absence





seizures and





myoclonic seizures





Second-line treatment





for partial seizures





and infantile spasms.





Intravenous use in





status epilepticus



Lamotrigine
Sodium
Partial seizures




channel
Tonic-clonic





Seizures associated





with Lennox-





Gastaut syndrome



Ethosuximide
Calcium
Absence




channel
seizures



Topiramate
GABA/Sodium
Seizures associated




channel
with Lennox-





Gastaut syndrome



Zonisamide
GABA/Calcium
Adjunctive therapy in




Sodium
adults with partial-




channel
onset seizures





Infantile spasm





Mixed seizure





Lennox-Gastaut





syndrome





Myoclonic





Generalised tonic-





clonic seizure



Levetiracetam
Calcium
Partial seizures




channel
Adjunctive therapy





for partial,





myoclonic and





tonic-clonic





seizures



Clonazepam
GABA
Typical and




receptor
atypical absences




agonism
Infantile myoclonic





Myoclonic seizures





Akinetic seizures



Rufinamide
Sodium
Adjunctive treatment




channel
of partial seizures





associated with





Lennox-Gastaut





syndrome
















TABLE 3







Examples of AEDs used specifically in childhood epilepsy











AED
Mechanism
Indication






Clobazam
GABA
Adjunctive therapy




receptor
in complex




agonism
partial seizures





Status epilepticus





Myoclonic





Myoclonic-absent





Simple partial





Complex partial





Absence seizures





Lennox-Gastaut





syndrome



Stiripentol
GABA
Severe myoclonic




receptor
epilepsy in




agonism
infancy





(Dravet syndrome)









The present invention demonstrates that patients with treatment-resistant childhood onset epilepsy have a poor response rate for seizure reduction when treated with the anti-epileptic drug stiripentol. Furthermore, the efficacy of the treatment with stiripentol is found to be reduced further when combined with the anti-epileptic drug clobazam. However, surprisingly when the cannabinoid cannabidiol (CBD) is provided in combination with either stiripentol or stiripentol and clobazam there is a significant and beneficial increase in the response rate in the ability of the drugs to reduce seizures.


Such an increase in efficacy by the addition of CBD is unexpected, particularly given the data that demonstrates a decrease in efficacy when the stiripentol is combined with clobazam.


BRIEF SUMMARY OF THE DISCLOSURE

In accordance with a first aspect of the present invention there is provided cannabidiol (CBD) for use in the reduction of seizures in treatment-resistant epilepsy, wherein the CBD is administered in combination with an anti-epileptic drug (AED) that works via GABA receptor agonism.


Preferably the AED that works via GABA receptor agonism is stiripentol.


In a further embodiment the CBD is administered in combination with stiripentol and clobazam.


Preferably the CBD is in the form of a highly purified extract of cannabis which comprises at least 98% (w/w) CBD and comprises less than 0.15% THC and up to 1% CBDV.


Alternatively, the CBD is present as a synthetic compound.


Preferably the dose of CBD is below 50 mg/kg/day. More preferably wherein the dose of CBD is greater than 10 mg/kg/day, more preferably the dose of CBD is greater than 20 mg/kg/day.


Preferably the treatment-resistant epilepsy is Lennox-Gastaut Syndrome or Dravet Syndrome.


In accordance with a second aspect of the present invention there is provided a method of treating treatment-resistant epilepsy, wherein the CBD is administered to an individual in need thereof in combination with stiripentol (STP).


Preferably the individual is a human.


DEFINITIONS

Definitions of some of the terms used to describe the invention are detailed below:


The cannabinoids described in the present application are listed below along with their standard abbreviations.









TABLE 4





Cannabinoids and their abbreviations

















CBD
Cannabidiol


embedded image







CBDA
Cannabidiolic acid


embedded image







CBDV
Cannabidivarin


embedded image







CBDVA
Cannabidivarinic acid


embedded image







THC
Tetrahydrocannabinol


embedded image











The table above is not exhaustive and merely details the cannabinoids which are identified in the present application for reference. So far over 60 different cannabinoids have been identified and 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).


“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.


“Highly purified cannabinoid extracts” 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 substantially removed, such that the highly purified cannabinoid is greater than or equal to 98% (w/w) pure.


“Synthetic cannabinoids” are compounds that have a cannabinoid or cannabinoid-like structure and are manufactured using chemical means rather than by the plant.


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.


“Treatment-resistant epilepsy” (TRE) or “intractable epilepsy” is defined as per the ILAE guidance of 2009 as epilepsy that is not adequately controlled by trials of one or more AED. Treatment resistant epilepsies such as Dravet syndrome or Lennox-Gastaut syndrome are difficult to treat childhood epilepsies. Often the treatment of seizures in patients with these syndromes involves the use of adjunctive therapy, e.g., treatment with more than one anti-epileptic drugs concurrently.


“Childhood epilepsy” refers to the many different syndromes and genetic mutations that can occur to cause epilepsy in childhood. Examples of some of these are as follows: Dravet Syndrome; Myoclonic-Absence Epilepsy; Lennox-Gastaut syndrome; Generalized Epilepsy of unknown origin; CDKL5 mutation; Aicardi syndrome; tuberous sclerosis complex;


bilateral polymicrogyria; Dup15q; SNAP25; and febrile infection related epilepsy syndrome (FIRES); benign rolandic epilepsy; juvenile myoclonic epilepsy; infantile spasm (West syndrome); and Landau-Kleffner syndrome. The list above is non-exhaustive as many different childhood epilepsies exist.







DETAILED DESCRIPTION
PREPARATION OF HIGHLY PURIFIED CBD EXTRACT

The following describes the production of the highly-purified (>98% w/w) cannabidiol extract which has a known and constant composition was used in the Examples below.


In summary the drug substance used is a liquid carbon dioxide extract of high-CBD containing chemotypes of Cannibis stavia 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 98% 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 5 below.









TABLE 5







Composition of highly purified CBD extract










Cannabinoid
Concentration






CBD
  >98% w/w



CBDA
NMT 0.15% w/w 



CBDV
NMT 1.0% w/w



Δ9 THC
NMT 0.15% w/w 



CBD-C4
NMT 0.5% w/w





>—greater than


NMT—not more than






EXAMPLE 1
DRUG-DRUG INTERACTION BETWEEN CANNABIDIOL (CBD) AND STIRIPENTOL (STP) DURING A CLINICAL TRIAL

The efficacy of CBD for the adjunctive treatment of seizures associated with Dravet syndrome was demonstrated in a single trial in patients aged 2 -18 years. Following completion of a 4-week baseline period, patients were randomized to receive either 20 mg/kg/day CBD (n=61) or placebo (n=59). CBD or placebo were added to their current anti-epileptic treatment which remained stable over the treatment period of the study.


All patients had a diagnosis of treatment-resistant Dravet syndrome and seizures were inadequately controlled with one or more concomitant anti-epileptic drugs (AEDs) with or without vagal nerve stimulation or ketogenic diet.


Seizure counts were reported daily via an Interactive Voice Response System.


Convulsive seizures were defined as all countable atonic, tonic, clonic, and tonic-clonic seizures. The primary efficacy end point was percent change from baseline in convulsive seizures.


At baseline, disease state characteristics were comparable between groups with 72.5% reporting an increase in seizure frequency with prior treatment and 15% never experiencing a reduction in seizure frequency with previous medications.


During a 4-week baseline period, patients were required to have at least 4 convulsive seizures (tonic-clonic, clonic, tonic or atonic) while on stable AED therapy. Patients had previously failed a median of 4 prior AEDs and 93% were taking 2 or more concomitant AEDs during the trial. The most commonly used concomitant AEDs (>25% of patients) were clobazam, valproate, stiripentol, levetiracetam, and topiramate.


The median percent change from baseline in reduction of convulsive seizures in Dravet Syndrome for the CBD 20 mg/kg/day group was statistically superior to placebo (p=0.0123).


Following the trial statistical analysis was performed on the various patient groups to determine whether there was an interaction between any of the concomitant AEDs that the patients were taking. Data for the interaction between CBD and stiripentol (STP) are described below.


Results


Table 6 below describes the percentage of patients that recorded a 50% reduction in convulsive seizures over the treatment period.









TABLE 6







Interaction between CBD and other AEDs









Percentage of patients with greater


Combination of AEDs
than 50% reduction in seizures





Stiripentol
14%


Stiripentol + Clobazam
 8%


Stiripentol + CBD
43%


Stiripentol + Clobazam + CBD
26%









As is shown, 14% of patients that were taking stiripentol and placebo experienced a greater than 50% reduction in the number of seizures from the number recorded during the 4 week baseline recording period.


Surprisingly, there was a reduction in efficacy in patients that were taking stiripentol and clobazam, where only 8% of these patients experienced a greater than 50% reduction in seizures from the baseline rate.


In the groups that were taking the test compound CBD, there was an increase in efficacy in both groups. It was found that 43% of patients that were on stiripentol and CBD obtained a greater than 50% reduction in convulsive seizures, whereas 26% of patients that were taking stiripentol, clobazam and CBD obtained a greater than 50% reduction in seizures.


Conclusions


The increase in efficacy brought about by the addition of CBD to the AED stiripentol provides a useful combination of therapy. Such an increase in efficacy was unexpected as when the stiripentol was combined with the


Furthermore, those patients that are already taking a combination of stiripentol and clobazam may benefit from the inclusion of CBD as an adjunct therapy as such an inclusion has been found to reduce the number of seizures.


As can be seen in Table 3 both stiripentol and clobazam are commonly used AEDs in childhood epilepsy syndromes, furthermore they both work via enhancement of gamma-aminobutyric acid (GABA) A-receptor agonism.


Stiripentol is a positive allosteric modulator of GABA-A receptors in the brain that enhances the opening duration of the channel by binding to a site different than the benzodiazepine binding site. Reduced synaptosomal uptake of GABA and/or inhibition of GABA transaminase may also explain the role of stiripentol in reducing seizures.


Clobazam binds at distinct binding sites at the post-synaptic GABA receptor. These GABA receptors are in various locations in the CNS (limbic, reticular formation) and clobazam increases the duration of time for which the receptor is open. As a result, hyper polarization and stabilization of the membrane occur as the post-synaptic inhibitory effect of GABA is enhanced.


Combination of CBD with AEDs that work as GABA receptor agonists may therefore prove to be of particular benefit in the treatment of childhood epilepsy syndromes.


EXAMPLE 2
DRUG-DRUG INTERACTION BETWEEN CANNABIDIOL (CBD) AND STIRIPENTOL (STP) IN HEALTHY VOLUNTEERS

As part of an open-label, fixed sequence, healthy volunteer trial the primary objective was to investigate the impact of CBD (750 mg twice daily) on the steady-state pharmacokinetics of stiripentol (STP) (750 mg) and the reciprocal effect on CBD, 7-hydroxy-cannabidiol (7-OH-CBD) and 7-carboxy-cannabidiol (7-COON-CBD).


Analyte plasma concentrations were determined using validated bioanalytical methods. A secondary objective was to evaluate the safety and tolerability of CBD when co-administered with STP.


When CBD was combined with STP (12 subjects) there was a 1.28-to 1.55-fold increase in exposure (Cmax and AUCtau). Co-administration of STP with CBD had no effect on CBD exposure; however, STP reduced 7-OH-CBD and 7-COOH-CBD exposure by 29% and 13% respectively.


The most common adverse event (AE) was diarrhoea, none of the effects on analyte exposure observed were likely to be clinically relevant or correlated with incidence or severity of AEs.


Conclusions


The above data demonstrate that the combination of CBD with STP provides a safe and efficacious combination treatment option.

Claims
  • 1-12. (Canceled)
  • 13. A method of reducing seizures in treatment-resistant epilepsy comprising administering cannabidiol (CBD) in combination with at least one anti-epileptic drug (AED) that works via GABA receptor agonism.
  • 14. The method of claim 13, wherein the AED that works via GABA receptor agonism is stiripentol.
  • 15. The method of claim 13, wherein the AED that works via GABA receptor agonism is stiripentol and clobazam.
  • 16. The method of claim 13, wherein the CBD is in the form of a highly purified extract of cannabis which comprises at least 98% (w/w) CBD.
  • 17. The method of claim 16, wherein the highly purified extract comprises less than 0.15% THC.
  • 18. The method of claim 16, wherein the extract further comprises up to 1 CBDV.
  • 19. The method of claim 13, wherein the CBD is present as a synthetic compound.
  • 20. The method of claim 13, wherein CBD is administered at a dose below 50 mg/kg/day.
  • 21. The method of claim 13, wherein CBD is administered at a dose greater than 10 mg/kg/day.
  • 22. The method of claim 13, wherein CBD is administered at 20 mg/kg/day.
  • 23. The method of claim 13, wherein the treatment-resistant epilepsy is Lennox-Gastaut Syndrome or Dravet Syndrome.
  • 25. The method of claim 23, wherein the treatment-resistant epilepsy is Lennox-Gastaut Syndrome.
  • 26. The method of claim 13, wherein the treatment-resistant epilepsy is Dravet Syndrome.
Priority Claims (1)
Number Date Country Kind
1801158.5 Jan 2018 GB national
Continuations (1)
Number Date Country
Parent 16960665 Jul 2020 US
Child 17552487 US