Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS OF TREATING LENNOX-GASTAUT SYNDROME USING FENFLURAMINE
Document Type and Number:
WIPO Patent Application WO/2017/035267
Kind Code:
A1
Abstract:
A method of treating and/or preventing symptoms of Lennox-Gastaut Syndrome (LGS) also known as Lennox Syndrome in a patient such as a patient previously diagnosed with Lennox Syndrome, by administering an effective dose of fenfluramine or its pharmaceutically acceptable salt to that patient. Lennox Syndrome patients are treated at a preferred dose of less than about 2.0 to about 0.01 mg/kg/day.

Inventors:
FARR STEPHEN J (US)
GALER BRADLEY S (US)
Application Number:
PCT/US2016/048470
Publication Date:
March 02, 2017
Filing Date:
August 24, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ZOGENIX INT LTD (GB)
International Classes:
A61K31/135; A61K31/137
Domestic Patent References:
WO2014177676A12014-11-06
Foreign References:
US20140343162A12014-11-20
US20110212171A12011-09-01
US20060270611A12006-11-30
US20110092535A12011-04-21
Other References:
WIRRELL, EC ET AL.: "Stiripentol in Dravet syndrome: Results of a retrospective U.S. study", EPILEPSIA, vol. 54, no. 9, 2013, pages 1595 - 1604, XP055368544, ISSN: 0013-9580
NAITHANI, M ET AL.: "The Conventional Antiepileptic Drug Use When Compared to a Combination Therapy Regime in a Teaching Hospital in India", INTERNATIONAL JOURNAL OF PHARMA AND BIO SCIENCES, vol. 3, no. 1, 2012, pages B-191 - B-197, XP055368545, ISSN: 0975-6299
GASTAUT ET AL., J. OF AUTISM AND DEVELOPMENTAL DISORDERS, vol. 17, 1987, pages 3
ZHANG ET AL., PLOS ONE, vol. 10, 2015, pages 5
See also references of EP 3340971A4
Attorney, Agent or Firm:
BOZICEVIC, Karl (US)
Download PDF:
Claims:
CLAIMS

That which is claimed is:

1. A method of treating, and/or preventing symptoms of Lennox- Gastaut syndrome (Lennox Syndrome) in a patient diagnosed with Lennox- Gastaut syndrome comprising administering an effective dose of fenfluramine or a pharmaceutically acceptable salt thereof to the patient.

2. The method of claim 1, wherein the effective dose is less than about 10.0 mg/kg/day to about 0.01 mg/kg/day.

3 The method of any one of claims 1-2, wherein the effective dose is

administered in the form of one or more dosage forms for oral, injectable, transdermal, inhaled, nasal, rectal, vaginal or parenteral delivery.

4. The method of any one of claims 1-3, wherein the dosage form is an oral solution in an amount selected from the group consisting of 120 mg or less, 60 mg or less, and 30 mg or less.

5. The method as claimed in any of claims 1 through 4 wherein the dosage form consists essentially only of fenfluramine as the active ingredient.

6. The method as claimed in any one of claims 1-4, wherein the effective dose of fenfluramine or a pharmaceutically acceptable salt thereof is co-administered with one or more co-therapeutic agents and the co-therapeutic agents can be selected from the group consisting of carbamazepine, ethosuximide, fosphenytoin, lamotrigine, levetiracetam, phenobarbital, progabide, topiramate, stiripentol, valproic acid, valproate, verapamil, and benzodiazepines such as clobazam, clonazepam, diazepam, ethyl loflazepate, lorazepam, midazolam and a pharmaceutically acceptable salt or base thereof.

7. Use of fenfluramine or a pharmaceutically acceptable salt thereof for treating, preventing and/or ameliorating seizures in a patient diagnosed with Lennox- Gastaut syndrome wherein the fenfluramine is formulated with a pharmaceutically acceptable carrier and an effective dose is less than about 10.0 mg/kg/day to about 0.01 mg/kg/day.

8. The use of claim 7 wherein the effective dose is selected from the group consisting of 120 mg or less, 60 mg or less, and 30 mg or less, and wherein the effective dose is administered in a dosage form selected from the group consisting of forms for oral, injectable, transdermal, inhaled, nasal, rectal, vaginal and parenteral delivery.

9. The use as claimed in any of claims 7-10, wherein the fenfluramine is the only active ingredient administered to the patient.

10. The use of any one of claims 7-10, wherein the effective dose of fenfluramine or a pharmaceutically acceptable salt thereof is co-administered with one or more co- therapeutic agents selected from the group consisting of carbamazepine, ethosuximide, fosphenytoin, lamotrigine, levetiracetam, phenobarbital, topiramate, stiripentol, valproic acid, valproate, verapamil, and benzodiazepines such as clobazam, clonazepam, diazepam, lorazepam, and midazolam and a pharmaceutically acceptable salt or base thereof.

11. The use of claim 10 wherein the co-therapeutic agent is a combination of stiripentol, clobazam, and valproate.

12. The use of claim 11, wherein the co-administration of the stiripentol, clobazam and valproate increases fenfluramine blood levels by 100% or more relative to fenfluramine blood levels obtained in the absence of the co-administration of stiripentol, clobazam and valproate, and further wherein blood levels of a metabolized of fenfluramine are decreased relative to levels of a flenfluramine metabolite obtained in the absence of the co-administration of stiripentol, clobazam and valproate.

13. The use of claim 10 which use is in combination with one or more co- therapeutic agents which is an anticonvulsant.

14. A kit for treating a symptom of Lennox- Gastaut syndrome (LGS) in a patient diagnosed with LGS, comprising:

a container comprising a plurality of doses of a formulation comprising a pharmaceutically acceptable carrier and an active ingredient comprising fenfluramine;

instructions for treating the patient diagnosed with LGS by withdrawing the formulation from the container, and administering the formulation to the patient.

15. The kit as claimed in claim 14, wherein:

the formulation is an oral solution comprising 2.5 milligram of fenfluramine in each milliliter of liquid solution; and

the instructions indicate dosing the patient based on patient weight and volume of oral solution administered.

16. The kit as claimed in claim 14, wherein the formulation is a solid oral formulation selected from the group consisting of: a tablet, a disintegrating table, a capsule, a lozenge, and a sachet.

17. The kit as claimed in claim 14, wherein said formulation is provided in a transdermal patch.

Description:
Methods of Treating Lennox-Gastaut Syndrome using Fenfluramine

FIELD OF THE INVENTION

[0001] This invention relates generally to the field of methods of treatment and in particular, methods of treating human patients, and more particularly towards treating human patients diagnosed with Lennox-Gastaut Syndrome.

BACKGROUND OF THE INVENTION

[0002] This invention relates to the treatment of symptoms of Lennox-Gastaut

Syndrome ("LGS," sometimes referred to as "Lennox Syndrome") using an

amphetamine derivative, specifically fenfluramine.

[0003] Fenfluramine, i.e. 3-trifluoromethyl-N-ethylamphetamine is an amphetamine derivative having the structure:

Systematic (lUPAC) name

(RS)-N-ethyl- l-[3-(trifluoromethyl)phenyl]propan- 2-amine

[0004] Fenfluramine was first marketed in the US in 1973 and had been administered in combination with phentermine to prevent and treat obesity. However, in 1997, it was withdrawn from the US and global market as its use was associated with the onset of cardiac valve fibrosis and pulmonary hypertension. Subsequently, the drug was withdrawn from sale globally and is no longer indicated for use in any therapeutic area. Without being bound by theory, the adverse effects associated with the use of fenfluramine as an anorexic agent are thought to be attributable to the interaction of fenfluramine's major metabolite norfenfluramine with the 5-HT2B receptor, which is associated with heart valve hypertrophy.

[0005] Fenfluramine is metabolized in vivo into norfenfluramine by cytochrome P450 enzymes in the liver. Cytochrome P450 enzymes such as CYP2D6 and CYP1A2 are primarily responsible for the production of norfenfluramine from fenfluramine in humans. Such metabolism includes cleavage of an N-ethyl group to produce norfenfluramine as shown below.

fenfluramine norfenfluramine

[0006] Despite past cardiovascular safety concerns that arose when high doses of

fenfluramine were used for treatment of adult obesity, attempts have been made to identify further therapeutic uses for that product, while weighing the known

cardiovascular risks of fenfluramine against potential therapeutic benefits. One disorder for which new treatment options are sorely needed is epilepsy, and in particular, epilepsy syndromes which are refractory to known treatments. Epilepsy is a condition of the brain marked by a susceptibility to recurrent seizures. There are numerous causes of epilepsy including, but not limited to birth trauma, perinatal infection, anoxia, infectious diseases, ingestion of toxins, tumors of the brain, inherited disorders or degenerative disease, head injury or trauma, metabolic disorders, cerebrovascular accident and alcohol withdrawal.

[0007] Prior to the inventor's work, investigation of fenfluramine's efficacy in epilepsy patients, while showing some initial promise, was far from definitive, and shared a common paradigm, i.e., that fenfluramine's primary effects were on behaviors that caused or induced seizures, not treating or preventing the seizure itself. [0008] For example, Aicardi and Gastaut (New England Journal of Medicine (1985),

313: 1419 and Archives of Neurology (1988) 45:923-925) reported four cases of self- induced photosensitive seizures, i.e., seizures caused by patients purposely staring into bright lights or the sun, that responded to treatment with fenfluramine.

[0009] Clemens, in Epilepsy Research (1988) 2:340-343 reported a case study wherein a boy suffering pattern sensitivity-induced seizures that were resistant to anticonvulsive treatment was treated with fenfluramine to curb the patient' s compulsive seizure- inducing behavior. Fenfluramine reportedly successfully terminated these self-induced seizures and the author concluded that this was because fenfluramine blocked the seizure- sensitive triggering mechanism, i.e., not by treating the seizure itself.

[0010] In Neuropaediatrics, (1996); 27(4): 171-173, Boel and Casaer reported on a study on the effects of fenfluramine on children with refractory epilepsy, all of whom exhibited compulsive seizure-inducing behavior. They observed that when

fenfluramine was administered at a dose of 0.5 to 1 mg/kg/day, this resulted in a reduction in the number of seizures experienced by the patients, and concluded that "this drug could have significant anti-epileptic activity in a selected group of young patients with idiopathy or symptomatic generalized epilepsy, namely, children with self-induced seizures." The authors noted that "[i]t may well be that fenfluramine has no direct antiepileptic activity but acts through its effect on the compulsion to induce seizures." Hence the authors seemed to suggest that fenfluramine affected behavior and not the seizure itself.

[0011] In a letter to Epilepsia, published in that journal (Epilepsia, 43(2):205-206,

2002), Boel and Casaer commented that fenfluramine appeared to be of therapeutic benefit in patients with intractable epilepsy and self-induced seizures. However, the authors did not attribute fenfluramine's efficacy to generalized anti-seizure activity.

[0012] A large number of subtypes of epilepsy have been characterized, each with its own unique clinical symptoms, signs, and phenotype, underlying pathophysiology and distinct responses to different treatments. The most recent version, and the one that is widely accepted in the art, is that adopted by the International League Against Epilepsy's ("ILAE") Commission on Classification and Terminology [See e.g.,, Berg et ah, "Revised terminology and concepts for organization of seizures," Epilepsia, 51(4):676-685 (2010)]:

[0013] I. ELECTROCHEMICAL SYNDROMES (arranged by age of onset):

[0014] A. Neonatal period

[0015] 1. Benign familial neonatal epilepsy (BFNE)

[0016] 2. Early myoclonic encephalopathy (EME)

[0017] 3. Ohtahara syndrome

[0018] B. Infancy

[0019] 1. Epilepsy of infancy with migrating focal seizures

[0020] 2. West syndrome

[0021] 3. Myoclonic epilepsy in infancy (MEI)

[0022] 4. Benign infantile epilepsy

[0023] 5. Benign familial infantile epilepsy

[0024] 6. Dravet syndrome

[0025] 7. Myoclonic encephalopathy in non-progressive disorders

[0026] C. Childhood

[0027] 1. Febrile seizures plus (FS+) (can start in infancy)

[0028] 2. Panayiotopoulos syndrome

[0029] 3. Epilepsy with myoclonic atonic (previously astatic) seizures

[0030] 4. Benign epilepsy with centrotemporal spikes (BECTS)

[0031] 5. Autosomal-dominant nocturnal frontal lobe epilepsy (ADNFLE)

[0032] 6. Late onset childhood occipital epilepsy (Gastaut type)

[0033] 7. Epilepsy with myoclonic absences

[0034] 8. Lennox- Gastaut syndrome

[0035] 9. Epileptic encephalopathy with continuous spike-and-wave during

[0036] sleep (CSWS), also known as Electrical Status Epilepticus during

[0037] Slow Sleep (ESES)

[0038] 10. Landau-Kleffner syndrome (LKS) [0039] 11. Childhood absence epilepsy (CAE)

[0040] D. Adolescence- Adult

[0041] 1. Juvenile absence epilepsy (JAE)

[0042] 2. Juvenile myoclonic epilepsy (JME)

[0043] 3. Epilepsy with generalized tonic-clonic seizures alone

[0044] 4. Progressive myoclonus epilepsies (PME)

[0045] 5. Autosomal dominant epilepsy with auditory features (ADEAF)

[0046] 6. Other familial temporal lobe epilepsies

[0047] E. Less specific age relationship

[0048] 1. Familial focal epilepsy with variable foci (childhood to adult)

[0049] 2. Reflex epilepsies

[0050] II. DISTINCTIVE CONSTELLATIONS

[0051] A. Mesial temporal lobe epilepsy with hippocampal sclerosis (MTLE

HS)

[0052] B. Rasmussen syndrome

[0053] C. Gelastic seizures with hypothalamic hamartoma

[0054] D. Hemiconvulsion-hemiplegia-epilep s y

[0055] E. Epilepsies that do not fit into any of these diagnostic categories,

[0056] distinguished on the basis of

[0057] 1. Presumed cause (presence or absence of a known structural or metabolic condition)

[0058] 2. Primary mode of seizure onset (generalized vs. focal)

[0059] III. EPILEPSIES ATTRIBUTED TO AND ORGANIZED BY

[0060] STRUCTURAL-METABOLIC CAUSES

[0061] A. Malformations of cortical development (hemimegalencephaly,

[0062] heterotopias, etc.)

[0063] B. Neurocutaneous syndromes (tuberous sclerosis complex,

[0064] Sturge-Weber, etc.)

[0065] C. Tumor [0066] D. Infection

[0067] E. Trauma

[0068] IV. ANGIOMA

[0069] A. Perinatal insults

[0070] B. Stroke

[0071] C. Other causes

[0072] V. EPILEPSIES OF UNKNOWN CAUSE

[0073] VI. CONDITIONS WITH EPILEPTIC SEIZURES NOT TRADITIONALLY

DIAGNOSED AS FORMS OF EPILEPSY PER SE

[0074] A. Benign neonatal seizures (BNS)

[0075] B. Febrile seizures (FS)

[0076] Part V of the ILAE classification scheme underscores the fact that the list is far from complete, and that there are still subtypes of epilepsy that have not yet been fully characterized, or that remain unrecognized as distinct syndromes.

[0077] Those skilled in the art will recognize that different subtypes of epilepsy are triggered by different stimuli, are controlled by different biological pathways, and have different causes, whether genetic, environmental, and/or due to disease or injury of the brain. In other words, the skilled artisan will recognize that teachings relating to one epileptic subtype are most commonly not necessarily applicable to any other subtype. Of particular importance is the fact that there are a large number of compounds that are used to treat different types of epilepsy, and different epilepsy subtypes respond differently to different anticonvulsant drugs. That is, while a particular drug may be effective against one form of epilepsy, it may be wholly ineffective against others, or even contra-indicated due to exacerbation of symptoms, such as worsening the frequency and severity of the seizures. As a result, efficacy of a particular drug with respect to a particular type of epilepsy is wholly unpredictable, and the discovery that a particular drug is effective in treating in treating a type of epilepsy for which that drug was not previously known to be effective is nearly always surprising, even in cases where the drug is known to be effective against another epilepsy type.

[0078] Lennox-Gastaut syndrome (LGS) was first described in 1960, and named

for neurologists William G. Lennox (Boston, USA) and Henri Gastaut (Marseille, France). It is a difficult-to-treat form of childhood-onset epilepsy that most often appears between the second and sixth year of life, although it can occur at an earlier or later age. LGS is characterized by frequent seizures and different seizure types; it is typically accompanied by developmental delay and psychological and behavioral problems. In children, common causes of LGS include perinatal brain injury, brain malformations such as tuberous sclerosis or cortical dysplasia, central nervous system (CNS) infection, and degenerative or metabolic disorders of the nervous system.

[0079] Daily multiple seizures of different types are typical in LGS. Also typical is the broad range of seizures that can occur. The most common seizure types are tonic-axial, atonic, and absence seizures, but myoclonic, generalized tonic-clonic, and focal seizures can also occur in any LGS patient. Atonic, atypical absence, tonic, focal, and tonic-clonic seizures are also common. Additionally, many LGS patients will have status epilepticus, often of the nonconvulsive type, which is characterized by dizziness, apathy, and unresponsiveness. Further, most patients have atonic seizures, also called drop seizures, which cause their muscles to go limp and result in the patient suddenly and unexpectedly to fall to the ground, often causing significant injury, which is why patients often wear a helmet to prevent head injury.

[0080] In addition to daily multiple seizures of various types, children with LGS

frequently have arrested/slowed psycho-motor development and behavior disorders.

[0081] The syndrome is also characterized by a specific finding on

electroencephalogram (EEG), specifically an interictal (i.e., between-seizures) slow spike- wave complexes and fast activity during sleep. Diagnosis

[0082] LGS is a syndrome and hence its diagnosis is based on the presence of specific clinical symptoms, signs, and laboratory tests. LGS is typically identified by a triad of features including multiple types of seizures, mental retardation or regression and abnormal EEG with generalized slow spike and wave discharges. Physicians use EEG to assist in diagnosing LGS. Diagnosis may be difficult at the onset of the initial symptom(s) because the triad of features associated with LGS, such as tonic seizures, may not be fully established, and EEG during sleep is required to confirm the condition. Thus, even though there may be overlap in clinical presentation with other epilepsies, LGS is agreed to be a well-defined distinct diagnosis by both the

International League Against Epilepsy (ILAE), considered the world' s leading expert medical society on epilepsy, and the FDA.

[0083] The diagnosis of LGS is more obvious when the patient suffers frequent and manifold seizures, with the classic pattern on the electro-encephalogram (EEG), i.e., a slowed rhythm with Spike-wave-pattern, or with a multifocal and generalizing sharp- slow-wave-discharges at 1.5-2.5 Hz. During sleep, tonic patterns (fast activity) can often be seen.

[0084] General medical investigation usually reveals developmental delay and

cognitive deficiencies in children with LGS. These may precede development of seizures, or require up to two years after the seizures begin, in order to become apparent.

[0085] There may be multiple etiologies for LGS, including genetic, structural,

metabolic or unknown. Approximately one-quarter have no prior history of epilepsy, neurological abnormality or developmental delay prior to the onset of LGS symptoms. Underlying pathologies causing LGS may include encephalitis and/or meningitis, brain malformations (e.g. , cortical dysplasias), birth injury, hypoxia-ischemia injury, frontal lobe lesions, and trauma.

[0086] An important differential diagnosis is 'Pseudo-Lennox-Syndrome', also called atypical benign partial epilepsy of childhood, which differs from LGS, in that there are no tonic seizures; sleeping EEG provides the best basis for distinguishing between the two. In addition, 'Pseudo-Lennox-Syndrome' has an entirely different etiology and prognosis than LGS.

Treatment

[0087] The optimum treatment for Lennox- Gastaut syndrome has yet to be established.

Many different treatments are currently used in the treatment of this disorder and many more have been tried in the past, most often with little success.

[0088] A variety of therapeutic approaches are currently used in LGS, including

conventional antiepileptic medications, diet and surgery, however the evidence supporting these therapies is not robust and treatment remains most often ineffective. The use of several common first-line treatments is based on clinical experience or conventional wisdom; examples include broad spectrum anti-convulsant medications, such as valproic acid, and benzodiazepines, most often clonazepam and clobazam. A few drugs have been proven effective for some patients for certain seizure types by double-blind placebo-controlled studies; examples include clobazam, lamotrigine, topiramate, felbamate, and rufinamide, although most patients continue to have significant seizures even while taking these medications. Second-line medications currently in use, such as zonisamide, are prescribed based on results of some open- label uncontrolled studies. The ketogenic diet may be useful in some patients with LGS refractory to medical treatment. Surgical options for LGS include corpus callostomy (for drop attacks), vagus nerve stimulation, and focal cortical resection (in the presence of a single resectable lesion). However, it should be noted that significant improvement from any of these therapies alone or in combination is a rare occurrence.

[0089] Despite the severity of LGS's symptoms and the frequency with which it occurs

(it accounts for up to 10% of all childhood epilepsies), there is currently no standard evidence-based treatment for the disease. A comprehensive review of the literature [see Hancock EC & Cross JH, Treatment of Lennox- Gastaut syndrome (Review), published in The Cochrane Library 2013, Issue 2] discovered only nine randomized controlled trials which evaluated the pharmaceutical treatment of the syndrome. The authors concluded that there is a paucity of research and "... that no monotherapy (to date) has been shown to be highly effective in this syndrome." Id at page 12. The authors further concluded that "[t]he optimum treatment for LGS remains uncertain and no study to date has shown any one drug to be highly efficacious". Id at page 12.

[0090] There is accordingly a dire and long-felt need to provide an improved method for treating or preventing and/or ameliorating seizures experienced by sufferers of Lennox- Gastaut Syndrome.

SUMMARY OF THE INVENTION

[0091] According to a first aspect of the present invention, there is provided a method of treating and/or preventing one or more symptoms of Lennox-Gastaut Syndrome in a patient comprising administering an effective dose to a patient of fenfluramine alone or in combination with one or more drugs as described here.

[0092] According to a further aspect of the present invention, there is provided a

method of treating, preventing and/or ameliorating seizures in a patient diagnosed with Lennox-Gastaut Syndrome comprising administering an effective dose to a patient of fenfluramine alone or in combination with one or more drugs as described here.

[0093] According to a further aspect of the present invention, there is provided a

method of treating a patient that exhibits a mutation in one or more of a gene selected from the group consisting of SCN1A, SCN1B, SCN2A, SCN3A, SCN9A, GABRG2, GABRD and PCDH19 by administering to that patient an effective dose of

fenfluramine.

[0094] A still further aspect of this invention contemplates a method for stimulating one or more 5-HT receptors in the brain of a patient by administering an effective dose of fenfluramine or a pharmaceutically acceptable salt thereof to that patient. Illustrative one or more 5-HT receptors are selected from the group consisting of one or more of 5- HTi, 5-HTiA, 5-HTiB, 5-HTic, 5-HTID, 5-HTIE, 5-HTIF, 5-HT 2 , 5-HT 2 A, 5-HT 2 B, 5- HT 2 c, 5-HT 3 , 5-HT 4 , 5-HT 5 , 5-HT 5A , 5-HT 5B 5-HT 6 , and 5-HT 7 . In addition there may be non-5-HT binding in the brain including Sigma, Ml muscarinic, B-adrenergic.

[0095] Yet another aspect of the invention contemplates co-administration of an

effective dose of one or more co-therapeutic agents with the fenfluramine wherein the co-therapeutic agents can be selected from the group consisting of carbamazepine, ethosuximide, fosphenytoin, lamotrigine, levetiracetam, phenobarbital, progabide, topiramate, stiripentol, valproic acid, valproate, verapamil, and benzodiazepines such as clobazam, clonazepam, diazepam, ethyl loflazepate, lorazepam, midazolam. Use of a pharmaceutically acceptable salt or base of a co-therapeutic agent is also contemplated.

[0096] An aspect of the invention is a method of treating or preventing the symptoms of Lennox-Gastaut syndrome (LGS) in a patient diagnosed with LGS comprising administering an effective dose of fenfluramine or pharmaceutically acceptable salt to the patient, wherein the dose is administered in an amount in the range of from 10.0 mg/kg/day to about 0.01 mg/kg/day, or administered at 120 mg or less; or 60 mg or less or 30 mg or less and may be administered in the absence of the administration of any other pharmaceutically active compound.

[0097] In another aspect of the invention, the method is carried out wherein the

effective dose is administered in a form selected from the group consisting of oral, injectable, transdermal, buccal, inhaled, nasal, rectal, vaginal, or parental, and wherein the formulation is oral, the formulation may be liquid which may be a solution or a suspension may be present within a container closed with a cap connected to a syringe graduated to determine the volume extracted from the container wherein the volume extracted relates to the amount of fenfluramine in a given liquid volume of formulation e.g. one millimeter of formulation contains 2.5 mg of fenfluramine. In another aspect of the invention, the method is administered in a solid oral formulation in the form of a tablet, capsule, lozenge, or sachet.

[0098] The method may be carried out as a co treatment with a different

pharmaceutically active compound. The method may be carried out in a process wherein the patient is first then subjected to a series of tests to confirm diagnoses of LGS.

[0099] Another aspect of the invention is a kit for treating Lennox- Gastaut syndrome

(LGS) in a patient diagnosed with LGS wherein the kit comprises a formulation comprising a pharmaceutically acceptable carrier and an active ingredient comprising fenfluramine and instructions for treating a patient diagnosed with LGS by

admnistereing the formulation to the patient. In yet another aspect, wherein the fenfluramine is in an oral liquid or a solid oral dosage form or a transdermal patch; and the kit further comprises instructions for treating a patient diagnosed with LGS by administering the formulation to the patient.

[00100] In another aspect of the invention, the kit consists of an oral liquid formulation in a container and a syringe with instructions, wherein the concentration of the fenfluramine in the liquid is calibrated based on calibrations on the syringe and includes calibrations wherein a milliliter of solution equates to a known amount of fenfluramine such as O. lmg, 0.2mg etc., to 1.0 mg.

[00101] In another aspect of the invention, the kit includes instructions relating to

dosing the patient based on patient weight and volume of solution based on the concentration of fenfluramine in the solution.

[00102] Another aspect of the invention is a use of a fenfluramine composition in

treating and or preventing symptoms of Lennox- Gastaut syndrome (LGS) and a patient diagnosed with LGS which use may include placing the fenfluramine in a liquid solution and withdrawing that liquid solution into a graduated syringe.

[00103] These and other objects, advantages, and features of the invention will become apparent to those persons skilled in the art upon reading the details of the methods of treating symptoms of Lennox- Gastaut Syndrome as more fully described below. BRIEF DESCRIPTION OF THE DRAWINGS

[00104] The present disclosure is best understood from the following detailed description when read in conjunction with the accompanying drawings. Included in the drawings are the following figures.

[00105] Figure 1 is a table summarizing the procedures followed during each of the patient visits which are conducted over the course of the clinical trial described in Example 1.

[00106] Figure 2 is a flow chart illustrating the manner in which fenfluramine dosages are increased for non-responding patients over the course of the clinical trial.

DETAILED DESCRIPTION OF THE INVENTION

[00107] Before the present method, kits and formulations are described, it is to be

understood that this invention is not limited to particular embodiments described, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.

[00108] Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limits of that range is also specifically disclosed. Each smaller range between any stated value or intervening value in a stated range and any other stated or intervening value in that stated range is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included or excluded in the range, and each range where either, neither or both limits are included in the smaller ranges is also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention.

[00109] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, some potential and preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited. It is understood that the present disclosure supersedes any disclosure of an incorporated publication to the extent there is a contradiction.

[00110] It must be noted that as used herein and in the appended claims, the singular forms "a", "an", and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a seizure" includes a plurality of such seizures and reference to "the formulation" includes reference to one or more formulations and equivalents thereof known to those skilled in the art, and so forth.

[00111] The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided may be different from the actual publication dates which may need to be independently confirmed.

[00112] After extensive research, it has unexpectedly been found that fenfluramine can be used to treat, or at least minimize the effects of Lennox-Gastaut Syndrome.

[00113] To avoid doubt, the term "prevention" of seizures means the total or partial prevention (inhibition) of seizures. Ideally, the methods of the present invention result in a total prevention of seizures. However, the invention also encompasses methods in which the instances of seizures are decreased in frequency by at least 40%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, or at least 90%. In addition, the invention also encompasses methods in which the instances of seizures are decreased in duration or severity by at least 40%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, or at least 90%. [00114] Without being bound by theory, fenfluramine has been known to trigger the release of serotonin (5-HT) in the brain due to disruption of its vesicular storage and to inhibit serotonin reuptake. However, until the present invention was made, it was not known that fenfluramine's mechanism of action made it suitable for the treatment of Lennox- Gastaut syndrome (LGS). In fact, there are no scientific publications demonstrating or even hypothesizing that 5-HT abnormalities are a possible underlying pathophysiologic cause for LGS or are causally related to the associated seizures in this specific epilepsy condition. Furthermore, since there has been no scientific hypothesis relating serotonin abnormalities in LGS, there are no studies nor even individual case reports in the medical literature which describe attempts to treat LGS using

medications that interacts with serotonin. The lack of data or even speculation in the literature regarding the use of fenfluramine or serotonergic agents in general to treat LGS are facts that strongly support the unexpected nature of this invention: given that LGS is a devastating refractory epilepsy condition and the number of people affected, investigators would be strongly motivated to investigate any treatment they perceived as having any potential for efficacy.

[00115] Thus, according to a still further aspect of the present invention, there is

provided a method of stimulating one or more 5-HT receptors in the brain of a patient by administering an effective dose of fenfluramine to said patient, said one or more 5- HT receptors being selected from one or more of 5-HTi, 5-HTIA, 5-HTI B , 5-HTI C , 5- HTID, 5-HT iE, 5-HTiF, 5-HT 2 , 5-HT 2 A, 5-HT 2 B, 5-HT 2 C, 5-HT 3 , 5-HT 4 , 5-HTS, 5-HTSA, 5-HT 5B 5-HT 6 , and 5-HT 7 amongst others. In certain embodiments of this aspect of the invention, the patient has been diagnosed with Lennox-Gastaut Syndrome.

[00116] In embodiments of the invention, any effective dose of fenfluramine can be employed. However, surprisingly low doses of fenfluramine have been found by the inventors to be effective, particularly for inhibiting or eliminating seizures in Lennox- Gastaut Syndrome patients.

[00117] DOSE BY WEIGHT (MG/KG/DAY) Thus in some cases, in a preferred

embodiment of the invention, a daily dose of less than about 10 mg/kg/day, such as less than about 10 mg/kg/day, less than about 9 mg/kg/day, less than about 8 mg/kg/day, less than about 7 mg/kg/day, less than about 6 mg/kg/day, less than about 5 mg/kg/day, less than about 4 mg/kg/day, less than about 3.0 mg/kg/day, less than about 2.5 mg/kg/day, less than about 2.0 mg/kg/day, less than about 1.5 mg/kg/day, less than about 1.0 mg/kg/day, such as about 1.0 mg/kg/day, about 0.95 mg/kg/day, about 0.9 meg/kg/day, about 0.85 mg/kg/day, about 0.85 mg/kg/day, about 0.8 mg/kg/day, about 0.75 mg/kg/day, about 0.7 mg/kg/day, about 0.65 mg/kg/day, about 0.6 mg/kg/day, about 0.55 mg/kg/day, about 0.5 mg/kg/day, about 0.45 mg/kg/day, about 0.4 mg/kg/day, about 0.350 mg/kg/day, about 0.3 mg/kg/day, about 0.25 mg/kg/day, about 0.2 mg/kg/day, about 0.15 mg/kg/day to about 0.1 mg/kg/day, about 0.075 mg/kg/day, about 0.05 mg/kg/day, about 0.025 mg/kg/day, about 0.0225 mg/kg/day, about 0.02 mg/kg/day, about 0.0175 mg/kg/day, about 0.015 mg/kg/day, about 0.0125 mg/kg/day, or about 0.01 mg/kg/day is employed. .

[00118] Put differently, a preferred dose is less than about 10 to about 0.01 mg/kg/day.

In some cases the dose is less than about 10.0 mg/kg/day to about 0.01 mg/kg/day, such as less than about 5.0 mg/kg/day to about 0.01 mg/kg/day, less than about 4.5 mg/kg/day to about 0.01 mg/kg/day, less than about 4.0 mg/kg/day to about 0.01 mg/kg/day, less than about 3.5 mg/kg/day to about 0.01 mg/kg/day, less than about 3.0 mg/kg/day to about 0.01 mg/kg/day, less than about 2.5 mg/kg/day to about 0.01 mg/kg/day, less than about 2.0 mg/kg/day to about 0.01 mg/kg/day, less than about 1.5 mg/kg/day to about 0.01 mg/kg/day, or less than about 1.0 mg/kg/day to

O.Olmg/kg/day, such as less than about 0.9 mg/kg/day, less than about 0.8 mg/kg/day, less than about less than about 0.7 mg/kg/day, less than about 0.6 mg/kg/day to about 0.01 mg/kg/day, less than about 0.5 mg/kg/day to about 0.01 mg/kg/day, less than about 0.4 mg/kg/day to about 0.01 mg/kg/day, less than about 0.3 mg/kg/day to about 0.01 mg/kg/day, or less than about.0.2 mg/kg/day to about 0.01 mg/kg/day.

[00119] As indicated above the dosing is based on the weight of the patient. However, for convenience the dosing amounts may be preset such as in the amount of 1.0 mg, 2.5 mg, 5 mg, 10 mg, 15 mg, 20 mg, 30 mg, 40 mg, or 50 mg. In certain instances, the dosing amount may be preset such as in the amount of about 0.25 mg to about 5 mg, such as about 0.25 mg, about 0.5 mg, about 0.75 mg, about 1.0 mg, about 1.25 mg, about 1.5 mg, about 1.75 mg, about 2.0 mg, about 2.25 mg, about 2.5 mg, about 2.75 mg, about 3.0 mg, about 3.25 mg, about 3.5 mg, about 3.75 mg, about 4.0 mg, about 4.25 mg, about 4.5mg, about 4.75mg, or about 5.0mg.

[00120] In general the smallest dose which is effective should be used for the particular patient.

[00121] The dosing amounts described herein may be administered one or more times daily to provide for a daily dosing amount, such as once daily, twice daily, three times daily, or four or more times daily, etc.

[00122] In certain embodiments, the dosing amount is a daily dose of 30mg or less, such as 30mg, about 29mg, about 28mg, about 27mg, about 26mg, about 25mg, about 24mg, about 23mg, about 22mg, about 21mg, about 20mg, about 19mg, about 18mg, about 17mg, about 16mg, about 15mg, about 14mg, about 13mg, about 12mg, about l lmg, about lOmg, about 9mg, about 8mg, about 7mg, about 6mg, about 5mg, about 4mg, about 3mg, about 2mg, or about lmg. In general the smallest dose which is effective should be used for the particular patient. In some cases, the dose is generally well below the dosing used in weight loss.

[00123] ROUTES OF ADMINISTRATION The dose of fenfluramine administered according to the methods of the present invention can be administered systemically or locally. Methods of administration may include administration via enteral routes, such as oral, buccal, sublingual, and rectal; topical administration, such as transdermal and intradermal; and parenteral administration. Suitable parenteral routes include injection via a hypodermic needle or catheter, for example, intravenous, intramuscular, subcutaneous, intradermal, intraperitoneal, intraarterial, intraventricular, intrathecal, and intracameral injection and non-injection routes, such as intravaginal rectal, or nasal administration. In certain embodiments, it may be desirable to administer one or more compounds of the invention locally to the area in need of treatment. This may be achieved, for example, by local infusion during, topical application, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.

[00124] DOSAGE FORMS / ROUTE OF ADMIN The dose of fenfluramine

administered in the methods of the present invention can be formulated in any pharmaceutically acceptable dosage form including, but not limited to (a) oral dosage forms such as tablets including orally disintegrating tablets, capsules, and lozenges, oral solutions or syrups, oral emulsions, oral gels, oral films, buccal liquids, powder e.g. for suspension, and the like; (b) injectable dosage forms; (c) transdermal dosage forms such as transdermal patches, ointments, creams; (c) inhaled dosage forms; and/or (e) nasally, (f) rectally, (g) vaginally administered dosage forms.

[00125] DOSGE FORM / FREQUENCY OF ADMIN Such dosage forms can be

formulated for once a day administration, or for multiple daily administrations (e.g. 2, 3 or 4 times a day administration). Alternatively, for convenience, dosage forms can be formulated for less frequent administration (e.g., monthly, bi-weekly, weekly, every fourth day, every third day, or every second day), and formulations which facilitate extended release are known in the art.

[00126] DOSAGE FORMS / PREPARATION, COMPONENTS The dosage form of fenfluramine employed in the methods of the present invention can be prepared by combining fenfluramine or a pharmaceutically acceptable salt thereof with one or more pharmaceutically acceptable diluents, carriers, adjuvants, and the like in a manner known to those skilled in the art of pharmaceutical formulation.

[00127] ORAL DOSAGE FORMS / SUITABLE FORMULATION TYPES &

COMPONENTS THEREOF In some embodiments, formulations suitable for oral administration can include (a) liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, or saline; (b) capsules, sachets or tablets, each containing a predetermined amount of the active ingredient

(fenfluramine), as solids or granules; (c) suspensions in an appropriate liquid; and (d) suitable emulsions. Tablet forms can include one or more of lactose, mannitol, corn starch, potato starch, microcrystalline cellulose, acacia, gelatin, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible excipients. Lozenge forms can include the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles including the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such excipients as are described herein.

[00128] ORAL DOSAGE FORMS / EXCIPIENTS, For an oral solid pharmaceutical formulation, suitable excipients include pharmaceutical grades of carriers such as mannitol, lactose, glucose, sucrose, starch, cellulose, gelatin, magnesium stearate, sodium saccharine, and/or magnesium carbonate. For use in oral liquid formulations, the composition may be prepared as a solution, suspension, emulsion, or syrup, being supplied either in solid or liquid form suitable for hydration in an aqueous carrier, such as, for example, aqueous saline, aqueous dextrose, glycerol, or ethanol, preferably water or normal saline. If desired, the composition may also contain minor amounts of non-toxic auxiliary substances such as wetting agents, emulsifying agents, or buffers.

[00129] By way of illustration, the fenfluramine composition can be admixed with conventional pharmaceutically acceptable carriers and excipients (i.e., vehicles) and used in the form of aqueous solutions, tablets, capsules, elixirs, suspensions, syrups, wafers, and the like. Such pharmaceutical compositions contain, in certain

embodiments, from about 0.1% to about 90% by weight of the active compound, and more generally from about 1% to about 30% by weight of the active compound. The pharmaceutical compositions may contain common carriers and excipients, such as corn starch or gelatin, lactose, dextrose, sucrose, microcrystalline cellulose, kaolin, mannitol, dicalcium phosphate, sodium chloride, and alginic acid. Disintegrators commonly used in the formulations of this invention include croscarmellose, microcrystalline cellulose, corn starch, sodium starch glycolate and alginic acid. [00130] Formulations suitable for topical administration may be presented as creams, gels, pastes, or foams, containing, in addition to the active ingredient, such carriers as are appropriate. In some embodiments the topical formulation contains one or more components selected from a structuring agent, a thickener or gelling agent, and an emollient or lubricant. Frequently employed structuring agents include long chain alcohols, such as stearyl alcohol, and glyceryl ethers or esters and oligo(ethylene oxide) ethers or esters thereof. Thickeners and gelling agents include, for example, polymers of acrylic or methacrylic acid and esters thereof, polyacrylamides, and naturally occurring thickeners such as agar, carrageenan, gelatin, and guar gum. Examples of emollients include triglyceride esters, fatty acid esters and amides, waxes such as beeswax, spermaceti, or carnauba wax, phospholipids such as lecithin, and sterols and fatty acid esters thereof. The topical formulations may further include other

components, e.g., astringents, fragrances, pigments, skin penetration enhancing agents, sunscreens (e.g., sunblocking agents), etc.

[00131] Particular formulations of the invention are in an oral liquid form. The liquid can be a solution or suspension and may be an oral solution or syrup, which is included in a bottle with a syringe graduated in terms of milligram amounts which will be obtained in a given volume of solution. The liquid solution makes it possible to adjust the volume of solution for appropriate dosing of small children, who can be

administered fenfluramine in an amount anywhere from 1.25 mg to 30 mg and any amount between in 0.25 milligram, increments and thus administered in amounts of 1.25 mg, 1.5 mg, 1.75 mg, 2.0 mg, etc.

[00132] A specific aspect of the invention is a treatment carried out to relieve symptoms of Lennox-Gastaut by the administration of only fenfluramine. However, the fenfluramine may be co-administered with other known pharmaceutical drugs such as a co-therapeutic agent selected from the group consisting of carbamazepine,

ethosuximide, fosphenytoin, lamotrigine, levetiracetam, phenobarbital, progabide, topiramate, stiripentol, valproic acid, valproate, verapamil, and benzodiazepines such as clobazam, clonazepam, diazepam, ethyl loflazepate, lorazepam, midazolam and a pharmaceutically acceptable salt or base thereof.

[00133] The co-therapeutic agents have recommended dosing amounts. Those

recommended dosing amounts are provided within the most current version of the Physician's Desk Reference (PDR) or htip://emedicine.med scape.com/ both of which are incorporated herein by reference specifically with respect to the co-therapeutic agents listed above and more specifically with respect to the dosing amounts recommended for those drugs.

[00134] In connection with the present invention, the co-therapeutic agent can be used in the recommended dosing amount or can be used in a range of from 100 th to 100 times 1/10 to 10 times 1/5 to 5 times 1/2 to twice the recommended dosing amount or any incremental 1/10 amount in between those ranges.

[00135] As a specific example of a combination of co-therapeutic agents with

fenfluramine, the co-therapeutic agent may be any one of or all three of stiripentol, clobazam, and valproate. The fenfluramine may be administered in the amount of 0.8mg/kg of patient body weight and co-administered with 3500mg of stiripentol, 20mg of clobazam, and 25mg per kg of valproate. Each of those amounts may be increased to twice, three times, five times, or ten times that amount or decreased by 10%, 50%, or 75%.

[00136] An aspect of the invention includes a kit for treating and or preventing

symptoms of LGS in a patient diagnosed with LGS, the kit comprising:

[00137] a container holding a liquid formulation of fenfluramine;

[00138] a dispensing device connected to the container and configured to

withdraw the liquid formulation from the container;

[00139] instructions for administering the liquid formulation to a patient in order to treat LGS.

[00140] In alternate embodiments, the dispensing device may be a syringe or graduated pipette useful for delivering varying doses of the fenfluramine liquid. In another embodiment, the dispensing device is a metered dosing device capable of dispensing a fixed volume of fenfluramine liquid. In one exemplary embodiment, the dose delivered by the metered dosing device is adjustable.

[00141] The formulation may be a solution or suspension and is prepared such that a given volume of the formulation contains a known amount of active fenfluramine.

[00142] For example, in one embodiment of this aspect, the dispensing device is a

syringe is graduated in one millimeter increments and the liquid fenfluramine formulation is characterized such that one millimeter in volume of formulation includes precisely one milligram of fenfluramine. In this manner, the patient may be correctly dosed with a desired milligram dosage of fenfluramine based on a volume of liquid formulation administered to the patient orally.

[00143] In alternate embodiments, the dispenser is a syringe connected to the container and configured to withdraw the liquid formulation from the container, wherein the syringe is marked with levels of graduation noting volume of formulation withdrawn, or a metered dose dispenser for delivering a predetermined volume of the formulation to said patient, or a metered dispensing device calibrated to deliver a predetermined volume of the liquid, permitting convenient, consistent, and accurate dosing.

[00144] In a method of the present invention, fenfluramine can be employed as a

monotherapy in the treatment of Lennox- Gastaut Syndrome. Alternatively,

fenfluramine can be co-administered in combination with one or more

pharmaceutically active agents, which may be provided together with the fenfluramine in a single dosage formulation, or separately, in one or more separate pharmaceutical dosage formulations. Where separate dosage formulations are used, the subject composition and ore or more additional agents can be administered concurrently, or at separately staggered times, i.e., sequentially.

[00145] In one embodiment, the agents are co-therapeutic agents, such as

anticonvulsants. Preferred co-therapeutic agents can be selected from the group consisting of carbamazepine, ethosuximide, fosphenytoin, lamotrigine, levetiracetam, phenobarbital, progabide, topiramate, stiripentol, valproic acid, valproate, verapamil, and benzodiazepines such as clobazam, clonazepam, diazepam, ethyl loflazepate, lorazepam, midazolam. Use of a pharmaceutically acceptable salt of a co-therapeutic agent is also contemplated.

[00146] Fenfluramine can be administered in the form of the free base, or in the form of a pharmaceutically acceptable salt, for example selected from the group consisting of hydrochloride, hydrobromide, hydroiodide, maleate, sulphate, tartrate, acetate, citrate, tosylate, succinate, mesylate and besylate. Further illustrative pharmaceutically acceptable salts can be found in Berge et al., J. Pharm Sci. (1977) 68(1): 1-19.

[00147] Fenfluramine for use in the methods of the present invention may be produced according to any pharmaceutically acceptable process known to those skilled in the art. Examples of processes for synthesizing fenfluramine are provided in the following documents: GB 1413070, GB 1413078 and EP441160.

[00148] The dose of fenfluramine to be used in a method of the present invention can be provided in the form of a kit, including instructions for using the dose in one or more of the methods of the present invention. In certain embodiments, the kit can

additionally comprise a dosage form comprising one or more co-therapeutic agents.

[00149] A method of the present invention can be practiced on any appropriately

diagnosed patient. In alternate exemplary embodiments of the present invention, the patient is aged about 18 or less, about 16 or less, about 14 or less, about 12 or less, about 10 or less, about 8 or less, about 6 or less or about 4 or less to about 0 months or more, about 1 month or more, about 2 months or more, about 4 months or more, about 6 months or more or about 1 year or more. Thus, in this embodiment, the diagnosed patient is about one month old to about 18 years old when treated.

[00150] The invention is further illustrated in the following Examples.

EXAMPLE 1

ADD-ON THERAPY WITH LOW DOSE FENFLURAMINE (FFA) IN LENNOX-GASTAUT

[00151] The efficacy of fenfluramine as an add-on treatments in Lennox-Gastaut

patients is studied in a Phase 2 Clinical Trial. The study protocol is described and preliminary results are presented here. Trial Objectives and Design

[00152] An open-label, non-placebo controlled add-on study was designed to assess the efficacy and safety of low-dose add-on fenfluramine across a range of fenfluramine doses (0.2, 0.4, 0.8 mg/kg/day, to a maximum of 30 mg/day). The trial was conducted over a 20 week period, with responders eligible for follow-on treatment, with follow- up appointments at three month intervals.

Inclusion and Exclusion criteria

[00153] Patients were recruited from childhood epilepsy clinics in Leuven and Antwerp,

Belgium, and selected for inclusion in the study according to criteria comprising a combination of age, physical and psychological characteristics, and resistance to treatment with conventional therapies. Details of selection criteria are provided below.

MINIMUM Multiple seizure types;

REQUIREMENTS: Tonic seizures present in all cases

Electro-clinical epilepsy

syndrome compatible with

Lennox- Gastaut syndrome

EEG showing slow spike saves and abnormal

background

Abnormal cognitive development

MRI compatible with Lennox-Gastaut epilepsy

(including no progressive disease)

Drug Resistant Seizures Patients received at least two anti-epileptic drugs

(AEDs), including VNS, during the 4 week period prior to inclusion

Eligible seizure types

• Tonic-clonic (GTC) - REQUIRED

• Tonic (TS)

• Atonic (AS)

Clearly recognizable focal (FS)

At least 4 documented seizures during 4 week period prior to inclusion

Minimum of 4 seizures in at least 2 separate weeks

Age Between 3 and 18 years [00154] Once enrolled, subjects are removed from the study in cases of serious adverse events, non-compliance, or lack of efficacy. Treatment is also stopped in the event of increased severity and frequency of seizures after discussion with the principle investigator; cardiac abnormalities (specifically, valvular problems), and/or adverse events (specifically, SAE, SAR or SUSAR) after discussion with the principle investigator. Patients may also withdraw voluntarily. Upon withdrawing, a safety examination (i.e., blood sampling and cardiac ultrasound) is performed and

fenfluramine use is tapered for one week at 50% of end dosage and then withdrawn completely.

Trial Protocol

[00155] Study duration is 20 weeks, ending at week 20 after inclusion of 20 patients, i.e., 20 weeks following the enrollment of the 20 th patient. Patients who respond to treatment are enrolled in a follow-on study and assessed on an ongoing basis.

[00156] Patients are examined during six clinical visits (VI through V6) scheduled at four week intervals, ending with V6 at week 20. Thereafter, responders continue in a follow-up study, with visits scheduled every three months. At each visit, endpoints and safety criteria are assessed, and dosages adjusted as necessary.

[00157] During the study period, patients are prohibited from receiving certain

medications and foods: (1) felbamate is prohibited as a concomitant medication unless the following criteria are met: the patient has been treated for at least 18 months prior to screening; has stable liver function and hematology laboratory tests, and the dose is expected to remain constant throughout the study; (2) dugs that interact with central serotonin, including imipramine, monoamine oxidase inhibitors, SSRIs, SNRIs, or vortioxetine; and (3) drugs or foods that potentially interact with fenfluramine via the CYP-2D6, CYPD-3A4, and/or CYP-2B6 pathways, except for pre-approved short-term use where required by medical necessity. Pregnancy testing, use of birth control, and breast feeding restrictions were also required during the study period and for the duration of subsequent fenfluramine treatment. [00158] An abbreviated trial flowchart appears in Table 1, shown in Figure 1. At VI, inclusion and exclusion criteria are assessed, clinical diagnosis confirmed, and the following information is collected: baseline demographics, pre -baseline seizure counts, current treatment regimens (both AEDs and VNS), and sleep quality. In addition, safety blood samples are collected, blood levels of anti-epileptic drugs ("AEDs") are determined, and cardiac function is evaluated using ultrasound imaging and EKG traces. Urine pregnancy tests in female subjects of childbearing potential are also done, and the patient's quality of life is assessed by means of clinical global impressions (both parents and physician) and sleep quality. Patients meeting entry criteria are enrolled and begin a prospective baseline period.

[00159] Add-on treatment is begun at V2. Participants being receiving an initial

fenfluramine dose of 0.2 mg/kg/day. Endpoints and safety criteria are assessed (seizure counting, adverse effects, pregnancy testing, and quality of life indicators (CGI, sleep scale)). Additional safety blood samples, AED blood levels and cardiac evaluation are done only if clinically indicated. A seizure diary and medication are dispensed.

[00160] At each of V3-V5, endpoints and safety criteria are again assessed. Dosages of non-responders are adjusted according to trial protocol. A schema for dose escalation is shown in Figure 2.

[00161] At V6, endpoints and safety criteria are again assessed. Safety blood samples,

AED blood levels and cardiac evaluation are repeated. Responders may enter the follow-up study. Non-responders begin FFA tapering, receiving 50% of the end dosage for 1 week and then discontinuing treatment.

[00162] Follow-up study: Responders at 20 weeks continue in a follow-up study, with visits scheduled at 3 month intervals. Patients receive a starting dose equal to the dosage received at week 20. At each visit, endpoints and safety criteria are assessed (seizure counting, current treatment, adverse effects, quality of life indicators (CGI, sleep scale)), pregnancy testing is performed, safety blood samples are collected, blood level AEDs are determined and cardiac function is evaluated using EKG and cardiac ultrasound. Dosages may be increased as necessary, up to a maximum of 30 mg/day. Follow up ends when fenfluramine becomes available as a regular treatment or at the election of the patient and/or treating physician if serious side effects occur and/or the drug is no longer effective.

Materials and Methods

[00163] Ethics and regulatory approvals: Trial conduct complies with the most recent version of the principles of the Declaration of Helsinki, the principles of GCP, and in accordance with all applicable regulatory requirements. The study protocol and related documents is subject to ethical review by all requisite authorities. Participants have given written informed consent prior to their enrollment and participation in compliance with all applicable laws, regulations and ethical guidelines as required, and ICFs are retained at participating trial sites in accordance with all applicable regulatory agencies and laws. All information and data related to the Study and disclosed to the Participating Site and/or Study Investigator are treated as confidential and will not be disclosed to third parties or used for any purpose other than the performance of the study. Data collection, processing and disclosure of personal data is subject to compliance with applicable personal data protections and personal data processing requirements.

[00164] Fenfluramine: Oral fenfluramine solution (2.5 mg/ml or 5 mg/ml) is provided by Zogenix Pharma. Starting dosage is 0.2 mg/kg/day BID; second step at 0.4 mg/kg/day BID; maximum dosage at 0.8 mg/kg/day BID or 30 mg/day BID, whichever is less. The drug is dispensed by Zogenix Pharma. Labeled bottles containing the oral fenfluramine suspension is given to patients and controlled at each visit. Bottle labels are kept in individual patient files. Calculation of bottle number and control of labels are done at the trial's conclusion. Patient compliance is assessed by control of oral solution quantity at each visit and collection of seizure diary with notification of drug intake.

[00165] Concomitant treatment: Lennox- Gastaut patients participating in the study receive concomitant treatment with two or more anti-epileptic drugs commonly used in the treatment of the disorder. The drug regimen is unchanged during baseline (the period from VI to V2) and the full trial period (V2 to V6).

[00166] Laboratory tests: Blood analysis and urine pregnancy tests are done in central lab at UZ Leuven. Safety blood samples are tested for hemogram, electrolytes, liver function (SGOT, SGPT, LDH, PT) and kidney function (urea, creatinine)). AED blood level determination is limited to phenytoin, phenobarbital, carbamazepine, and valproate.

[00167] Safety assessment: Treatment safety is assessed using a combination of

physical examination, blood testing, cardiac evaluation, and adverse event reporting. With respect to adverse event reporting, reporting is not required for expected AEs, moderate weight loss and decrease of appetite with no significant weight loss (<P3).

[00168] Data Handling and Statistical Analysis: Data is coded and is protected from disclosure outside of research teams according to the terms of the research protocol and the informed consent document. Subjects' names or other identifiers must be stored separately ("site file") from their research data and replaced with a unique code to create a new identify for the subject. Coded data are not anonymous. Data is collected in standardized CRF.

[00169] A priori data on possible efficacy is unavailable. Sample size is set at 20. The study is not randomized. Descriptive analysis of outcome parameters is done at weeks 8, 12, 16 and 20. All included subjects are counted for analysis. Reasons for withdrawal are documented.

Interim Results

[00170] At the time results were reported, a total of 13 patients had been recruited and

12 enrolled. 9 patients were receiving treatment; 3 others were within the baseline period (VI to V2).

[00171] None of the 9 patients receiving fenfluramine reported serious side effects. 3 patients receiving 0.2 mg/kg/day showed significant decreases in total seizure frequency. [00172] The early interim data suggests that low-dose fenfluramine used as an add-on treatment is effective in reducing seizure frequency in Lennox-Gastaut patients.

[00173] The preceding merely illustrates the principles of the invention. It will be

appreciated that those skilled in the art will be able to devise various arrangements which, although not explicitly described or shown herein, embody the principles of the invention and are included within its spirit and scope. Furthermore, all examples and conditional language recited herein are principally intended to aid the reader in understanding the principles of the invention and the concepts contributed by the inventors to furthering the art, and are to be construed as being without limitation to such specifically recited examples and conditions. Moreover, all statements herein reciting principles, aspects, and embodiments of the invention as well as specific examples thereof, are intended to encompass both structural and functional equivalents thereof. Additionally, it is intended that such equivalents include both currently known equivalents and equivalents developed in the future, i.e., any elements developed that perform the same function, regardless of structure. The scope of the present invention, therefore, is not intended to be limited to the exemplary embodiments shown and described herein. Rather, the scope and spirit of present invention is embodied by the appended claims.