throbber
1
`
`Quality aspects
`
`SCIENTIFIC DISCUSSION
`
`Introduction
`1.1
`Sodium oxybate is a simple molecule, the sodium salt of gamma-hydroxybutyric acid (GHB) and is
`presented in the form of a stabilised oral solution, 500mg/ml
`
`1.2 Active Substance
`Sodium oxybate is the Common Name of the substance butanoic acid 4-hydroxy-monosodium salt. At
`the time of writing this report there is no INN for this substance.
`
`1.2.1 Manufacture
`The manufacturing process is very simple and is basically a one-step hydrolysis of gamma
`butyrolactone under alkaline conditions with sodium hydroxide.
`Starting materials and critical steps are well defined; there are no intermediates.
`The active substance is obtained as a white solid which is dried, ‘de-lumped’ and packed.
`The characterisation of the substance arising from the documented method of synthesis confirms that it
`is indeed sodium oxybate and this has been done by the usual range of spectroscopic methods
`including UV, IR & NMR together with elemental analysis and pKa determination.
`Since sodium oxybate is to be given in solution, polymorphism has not been investigated
`
`1.2.2 Specification
`The specification includes test for identification (IR, HPLC), assay (HPLC), related impurities
`(HPLC) together with tests for water content (KF) residual solvents (GC) and heavy metals, all
`performed by validated methods. It is not necessary to control solid state properties.
`The impurities include a number of named impurities and one un-named impurity, the levels of which
`have all been qualified on a toxicological basis and are considered to present no unnecessary risk.
`Batch analyses (n = 45) from the site of manufacture defined in the dossier, demonstrate satisfactory
`compliance with the agreed specification and indicate good uniformity.
`
`1.2.3 Stability
`In addition to forced degradation studies in the solid state and in solution, stability studies have been
`performed on 6 batches of sodium oxybate under ICH conditions, accelerated and long term. No
`significant negative trends or out of specification results were observed in the formal ICH stability
`investigation, and on the basis of the accumulated results a satisfactory re-test period has been defined.
`
`1.3 Medicinal Product
`
`1.3.1 Pharmaceutical Development
`The product is a simple aqueous solution which is stabilised with malic acid (hydroxysuccinic acid)
`and adjusted to the pH of maximum stability. From a microbiological point of view, investigations on
`this formulation showed that the product also had intrinsic antimicrobial activity and passed the test
`for efficacy of antimicrobial preservatives; therefore it was considered unnecessary to include a
`preservative in the formulation.
`The product is presented as a plastic PET bottle with a child-resistant screw cap closure for the liquid,
`and in order to facilitate accurate dosage a separate dispensing or dosing system is attached at time of
`first use. This consists of a plastic adaptor to fit into the bottle, leading to a syringe dispenser allowing
`the patient to withdraw the accurate dose. In addition, two plastic dosing cups are provided with child-
`resistant closures.
`
`PAR1021
`IPR of U.S. Patent No. 8,772,306
`Page 1 of 30
`
`

`
`1.3.2 Manufacture of the Product
`The active substance is dissolved in purified water and the pH adjusted with malic acid before dilution
`with purified water, filtration, and bottling. The validation of this simple scheme was not
`problematical.
`
`1.3.3 Product Specification
`The product release specification includes relevant tests and limits for physical examination, identity
`(HPLC & IR), assay (HPLC), impurities (degradation products, HPLC), volume in container,
`reproducibility of dosage, pH, microbiological attributes (PhEur), etc. Control of rheological
`properties is not considered necessary for a mobile liquid dosage form.
`Batch analyses (n = 18) indicate satisfactory compliance with the agreed specification and satisfactory
`product uniformity
`
`1.3.4 Stability of the Product
`In all, ten batches of product have been investigated for stability under ICH conditions, accelerated
`and long term, and in all cases the results support the shelflife and storage conditions as defined in the
`SPC.
`In addition, since this is a multidose product with a special adapter and dosing system, additional
`studies were performed with these plastic components in place, in order to mimic the in-use situation,
`and a suitable in-use shelflife has been defined.
`In general the studies show that the plastic adapter and dosing system is compatible with the solution
`and does not encourage degradation.
`Apart from the physical chemical analytical studies carried out during the stability investigations,
`microbiological studies were also performed with satisfactory results.
`
`1.4 Discussion on chemical, pharmaceutical and biological aspects
`The simple synthesis and manufacture of the product are described and controlled in a relevant
`manner, and the specifications of the active substance and medicinal product are considered to be
`relevant for a product of this type. The stability of the product has been well-investigated, both in the
`unopened form, and with the adaptor and dosing system in place during use.
`Satisfactory uniformity of dose has been demonstrated, and there are no unresolved quality issues that
`could have an impact on the benefit/risk balance for the patient.
`
`2
`
`Non-clinical aspects
`
`2.1
`
`Introduction
`
`2.2 Pharmacology
`
`2.2.1 Primary pharmacodynamics (in vitro/in vivo)
` Oxybate (GHB) is a metabolite of γ-aminobutyric acid (GABA) which is synthesised and
`accumulated by neurones in the brain. It is present at µM concentrations in all brain regions
`investigated as well as in several peripheral organs, particularly in the gastro-intestinal system.
`Neuronal depolarisation releases GHB into the extracellular space in a Ca2+-dependent manner. A
`family of GHB receptors in rat brain have been identified and cloned and most probably belong to the
`G-protein-coupled receptors. High-affinity receptors for GHB are present only in neurones, with a
`restricted specific distribution in the hippocampus, cortex and dopaminergic structures of rat brain.
`In general, stimulation of these receptors with low (physiological) amounts of GHB induces
`hyperpolarisation in dopaminergic structures with a reduction of dopamine release. However, in the
`hippocampus and frontal cortex, GHB seems to induce depolarisation with an accumulation of cGMP
`and an increase in inositol phosphate turnover. However, at higher (therapeutic) exposures, GHB
`
`Page 2/30
`
`PAR1021
`IPR of U.S. Patent No. 8,772,306
`Page 2 of 30
`
`

`
`receptors are saturated and probably de-sensitised and down-regulated. Such GHBergic potentiations
`induce dopaminergic hyperactivity, strong sedation with anaesthesia and EEG changes that are
`consistent with normal sleep and/or epileptic spikes.
`The pathogenesis of narcolepsy is still unknown, but an imbalance between monoamines and
`acetylcholine is generally accepted. Recent research has found a marked reduction of the neuropeptide
`hypocretin type 1 in the cerebrospinal fluid of a majority of patients and a global loss of hypocretins in
`post-mortem brain tissue of narcoleptic subjects. The hypocretins are synthesised by a small group of
`neurones predominantly located in the lateral hypothalamic and perifornical regions of the
`hypothalamus. The hypothalamic system directly and strongly innervates and potently excites
`noradrenergic, dopaminergic, serotoninergic, histaminergic and cholinergic neurones. The effect of
`GHB on this system has not been investigated. However, the available data indicate that its mode of
`action is likely to relate to non-specific dopaminergic stimulation rather than the hypocretin system.
`Formal nonclinical pharmacology studies to investigate the primary pharmacodynamics have not been
`conducted by the applicant, rather a comprehensive review of the scientific literature has been
`conducted. The publications included have been selected based on their relevance to the proposed
`indications, based on evidence of efficacy from early clinical studies. In addition, animal models of
`cataplexy and narcolepsy are continuing to be developed, but have not yet been fully validated. Little
`nonclinical information is available on its effects on narcolepsy in general, and cataplexy in particular.
`Available, directly relevant data, from the published literature, has been reviewed but the current
`understanding of the role of GHB in the CNS does not provide a mechanistic explanation of the
`positive clinical effects reported in the dossier. GHB had no effect on cataplexy in dogs with
`hereditary narcolepsy when administered as a single dose of 500 mg/kg i.v. or 50 mg/kg/day p.o. for 3
`consecutive days. However, although such dogs have a mutation of the type 2 hypocretin receptor, the
`clinical relevance of this model remains to be established. Moreover, a dose of 75 mg/kg/day for at
`least 14 days is required for efficacy in humans.
`Though the precise mode of action is unknown, the sedative properties of GHB and its effects on sleep
`may play a role in the efficacy observed in humans.
`Evidence from a human clinical study (Study OMC-SXB-20) where GHB was administered to
`narcoleptic patients and overnight polysomnograms (PSG) were recorded, suggests that GHB modifies
`sleep architecture, specifically a dose-related increase in Stage 3 & 4 slow wave sleep (SWS, delta
`sleep). The cause of human narcolepsy and cataplexy is, as yet, unknown. Recent evidence points to
`the loss of hypocretin-containing neurones, possibly due to autoimmune attack, as a likely cause
`(Scammell 2003). Hypocretin is a neurotransmitter that has roles amongst others, in sleep-wake
`regulation. Alterations in hypocretin neurotransmission have also been observed in mouse and dog
`models of narcolepsy, although no studies have been undertaken with GHB in these models. Animal
`models of cataplexy and narcolepsy are continuing to be developed (Gerashchenko et al, 2003), but
`the effects of GHB in these models, have yet to be investigated.
`
`2.2.2 Secondary pharmacodynamics
`Published literature reports are presented that discuss the potential for effects on the respiratory,
`cardiovascular, gastrointestinal, renal and endocrine function, together with relevant findings from the
`toxicology studies.
`GHB may increase growth hormone secretion, but this effect is inconsistent across species and dose
`levels. GHB has no other relevant secondary pharmacodynamic effects in animals.
`GHB consistently decreases respiration by effects on minute volume and respiratory rate, with
`younger animals being more susceptible to these effects. In halothane-anaesthetized rats, GHB (187.5-
`750 mg/kg i.p.) dose-dependently decreased basal minute volume and respiratory rate compared to
`pre-injection control, with a maximum decrease to about 60% of pre-injection values for each
`parameter at the highest dose of GHB (Hedner et al, 1980).
`Effects on cardiovascular parameters were also studied as part of the repeat dose toxicology studies in
`dogs, including heart rhythm and P-QRS-T complexes determined from ECGs, and there was no
`evidence of any dramatic changes in these parameters during the studies at doses up to 600 mg/kg/day
`(corresponding to male and female AUC0-24 of 3363.05 and 3631.35 µg·hr/mL and Cmax of 583.0
`
`Page 3/30
`
`PAR1021
`IPR of U.S. Patent No. 8,772,306
`Page 3 of 30
`
`

`
`and 726.7 µg/mL). Additionally, in several human clinical studies, there were no significant effects of
`GHB administration on ECGs. The applicant claims that given the relatively long established clinical
`use of GHB, as an anaesthetic and sedative and other uses such as for the treatment of alcohol
`withdrawal, the undesirable effects and risk potential of GHB are well known, and additional
`nonclinical safety pharmacology studies are not justified. Results of some studies indicate weak
`rewarding effects and possible development of tolerance in rats and mice, however there is no
`compelling evidence that GHB represents a significant drug dependence hazard. Interaction of GHB
`with ethanol and other central nervous system depressants generally result in greater central depressant
`effects than seen with either drug alone. Numerous case reports of GHB poisoning demonstrate that
`overdosing in humans is associated with many of the same signs and symptoms as in animals: a rapid
`onset of drowsiness, nausea, vomiting, myoclonic seizures, respiratory depression progressing to
`apnoea, and coma.
`
`2.2.3 Safety pharmacology
`The applicant has not conducted animal safety pharmacology studies. However, there is ample
`evidence in the published literature that GHB is a potent CNS depressant, may cause convulsions and
`potentially fatal respiratory depression and cardiac failure.
`On the basis of available information, the lack of conventional safety pharmacology studies is
`considered acceptable. However, since co-medication is probable, the effects of GHB on respiratory
`pattern in the presence of other CNS depressing agents like ethanol, and inhibitors of GHB
`metabolism
`like
`valproic
`acid
`and
`ethosuximide
`are mentioned
`in
`the
`SPC.
`A weak tolerance to GHB administration has been demonstrated in a number of specific animal
`behavioural studies and also the development of cross-tolerance between GHB and ethanol. Therefore,
`potentially, an acute toxic effect (e.g. acute respiratory depression) could be experienced after drug
`intake following a period of drug withdrawal, sufficient for the disappearance of tolerance. Caution is
`advised if treatment is re-started after discontinuation. (SPC, section 4.2). Clinical data (open label
`study OMC-GHB-3) have failed to show any major development of tolerance on efficacy and the
`AUC after 8-weeks compared to the first dose was not significantly increased (study OMC-SXB-10,
`see clinical section). However, as these clinical data are too limited to draw firm conclusions, the
`potential for development of tolerance, especially with concomitant intake of ethanol, cannot be
`excluded and is mentioned in the SPC.
`
`2.2.4 Pharmacodynamic drug interactions
`Formal studies of pharmacodynamic drug interactions have not been conducted. According to the
`published literature, concomitant administration of GHB and other CNS depressants (benzodiazepines,
`barbiturates, alcohol) results in an additive increase in sedation.
`
`2.3 Pharmacokinetics
`The applicant has not conducted animal PK studies, with the justification that the more relevant
`pharmacokinetic data are derived from human exposure. Some data have been compiled from a review
`of the published literature. Data on non-clinical absorption, distribution, metabolism, and excretion
`have been compiled from a review of the published literature
`
`2.3.1 Absorption- Bioavailability
`In the rat, oral bioavailability was about 50-80%. Kinetics was non-linear, with oral dose increments
`resulting in an under-proportional increase in Cmax and an over-proportional increase in the AUC. By
`contrast, i.v. administration resulted in an over-proportional increase in Cmax. Thus, both absorption
`from the gut and elimination may depend on saturable mechanisms. Saturable absorption from the gut
`was confirmed in an everted rat intestine model.
`
`2.3.2 Distribution
`Whole-body autoradiography following i.v. injection of 14C-GHB in mice showed a fairly uniform
`distribution pattern of radioactivity due to GHB and/or its metabolites. Shortly after injection, lower
`radioactivity was found in fatty tissues such as thymus, brown fat, and the white and grey brain matter
`
`Page 4/30
`
`PAR1021
`IPR of U.S. Patent No. 8,772,306
`Page 4 of 30
`
`

`
`than in other tissues, including plasma. However, by 30 minutes after injection, radioactivity was
`distributed throughout the body, including brain, skeletal muscle, myocardium, kidney, spleen, liver,
`lung, thymus, urinary bladder, stomach, intestines, and, in pregnant mice, the foetus. GHB distributed
`rapidly to the brain of rats, dogs and monkeys, producing brain concentrations several orders of
`magnitude above the physiological level. In the dog, the highest concentration was found in the white
`matter of the temporal lobe. There are no data on plasma protein binding in animals, but this is likely
`to be negligible.
`
`2.3.3 Metabolism (in vitro/in vivo)
`Metabolism of GHB is rapid and complete and proceeds via succinic semialdehyde, succinate and the
`Krebs cycle or through γ-hydroxybutyrate and β-oxidation.
`The potential for inhibition of CYP isozymes was tested in pooled human liver microsome fractions
`using standard markers for CYP1A2, 2C9, 2C19, 2D6, 2E1 and 3A. In all cases, the IC50 was > 3000
`µM (> 378 µg/ml). Since the average maximum human exposure is 142 µg/ml, GHB is not expected to
`show pharmacokinetic interactions with drugs metabolised by these isozymes. Non P450 mediated
`effects on GHB metabolism are unclear.
`A rat study found that co-administration of compounds stimulating or inhibiting GHB dehydrogenase
`were able to decrease or increase plasma levels of GHB by up to 1/3. The interactions resulting from
`the stimulation or inhibition of GHB dehydrogenase, namely with anticonvulsivant drugs and L-dopa
`are considered to be clinically relevant and are mentioned in the SPC.
`
`2.3.4 Excretion
`Clearance is predominantly by biotransformation, with limited amounts of unchanged drug recovered
`from the urine or faeces. Radiospirometric studies in rats showed that 14C-GHB was rapidly
`converted to exhaled CO2 and about 2/3 of the dose was excreted by respiration within 6 hours and an
`additional 10- 20% over the next 18 hours. After oral administration of 14C-labelled GHB
`(200 mg/kg) to rats, the urinary recovery over 48 hours was 5.5% of the radioactive dose, and only
`1.5% was recovered in the faeces. There are no data on the excretion of GHB in the milk of lactating
`animals. The proposed SPC contains an appropriate statement to this effect.
`T½ in rats following oral administration of a single dose of 200 mg/kg was 0.75 h for the α- and 2.68
`h for the β-phase. Similar T½ values were observed in dogs and monkeys. In rats, Cmax and AUC
`values tended to be higher in females than in males, whereas the opposite applied to dogs.
`The applicant has been asked to discuss the comparative pharmacokinetics in humans and
`experimental animals and the implications for a critical appraisal of the relevance of the main species
`used in the toxicity testing for human safety assessment. In summary, the rat and dog showed similar
`pharmacokinetic characteristics, although exposure measured as AUC was higher in human than in
`either rat or dog at the NOAEL. The exposures measured in the maximum tolerated dose toxicokinetic
`studies (conducted in support of mouse and rat carcinogenicity studies) were, however, greater than in
`human subjects (Cmax 2.60- and 2.76-fold; AUC 1.21- and 1.64-fold for mouse and rat, respectively).
`
`2.4 Toxicology
`All toxicology studies were conducted by the applicant with the exception of data from literature for
`single dose toxicity and carcinogenicity in mice.
`2.4.1 Single dose toxicity
`Formal single dose toxicity studies were not conducted. A review of published literature data
`identified a number of references providing LD50 values in several species. In the mouse, LD50
`values of 2960 – 3700 mg/kg following i.p. injection were identified; in the rat, LD50 values were
`9990 mg/kg following p.o. administration, and 1700 mg/kg following i.p. injection. In the rabbit and
`dog, LD50 values in excess of 1000 mg/kg were reported following i.v. administration, which could
`be increased to over 7000 mg/kg with artificial respiration without lethality.
`
`Page 5/30
`
`PAR1021
`IPR of U.S. Patent No. 8,772,306
`Page 5 of 30
`
`

`
`2.4.2 Repeat dose toxicity (with toxicokinetics)
`Repeat-dose toxicity studies comprised 3- and 6-month toxicity studies in rats and 3- and 12-month
`studies in dogs. Treatment-related clinical signs were mainly related to sedation, reduced food
`consumption and secondary changes in body weight, body weight gain and organ weights.
`In rats, the only treatment-related clinical chemistry changes were a slight reduction of serum albumin
`and WBC in rats that may have been related to changes in nutritional status. The lowest NOAEL value
`was 350 mg/kg/day in rats (AUC ≈200 µg.h/ml) based on bodyweight changes.
`In dogs, three repeat dose studies have been conducted, an initial rising dose study, followed by 90
`day and 52 week exposure studies. In the rising dose study single doses ranging from 150 – 1800
`mg/kg/day were investigated, followed by a 5 day continuous dosing phase at 600 mg/kg/day.
`Treatment related clinical signs included emesis following dosing at 600 mg/kg and 1200 mg/kg and
`emesis, hypersalivation, ataxia and hypoactivity following the 1800 mg/kg dose. Emesis and ataxia
`were also observed during the 600 mg/kg/day daily dosing phase. Plasma concentrations were
`observed to increase in approximate proportion to increasing dose, with emesis leading to decreased
`plasma concentrations. These signs subsided after the first few weeks of the study, and led to the
`decision to increase the high dose to 900 mg/kg/day at Week 32. Following this dose increase, similar
`clinical signs were noted in the 900 mg/kg/day dose group.
`There were no treatment-related clinical chemistry, ophthalmology or ECG findings. Necropsy
`findings included a dark area on the ileal mucosa of one animal, as a result of emesis during the study.
`A dose-dependent atrophy of the salivary and submucosal oesophageal glands was observed. Such
`changes are not uncommon with drugs acting on the parasympathetic nervous system. The NOAEL
`value was 150 mg/kg/day (AUC ≈300 µg.h/ml) based on bodyweight changes and salivary gland
`atrophy.
`The toxicokinetic parameters (mean values calculated from male and female data, since no apparent
`gender difference has been reported) at the NOAEL and/or NOELs in the repeat dose toxicology
`studies in rats and dogs are presented in the next table, together with pharmacokinetic parameters for
`humans from a study where the highest proposed dose of GHB was administered (2 x 4.5 g, 4 h apart,
`Study OMC-SXB-9).
`
`
`
`At the NOAEL observed in both rat repeat dose toxicology studies, mean AUC and Cmax values were
`approx 0.5 fold the values in humans. After the second 4.5 g dose in humans, the Cmax values for rats
`and humans were approximately the same. At the lowest NOEL for dogs, observed in the dog 52 week
`toxicology study (150 mg/kg/day), the exposure margin for AUC was approximately 0.5 fold the
`values observed in humans. The exposure margin for Cmax was approx 0.5 fold the value following
`the first dose in humans and approximately the same following the second dose in humans. Thus, with
`the exception of Cmax values following the second GHB dose in humans, exposure to GHB in both
`rats and dogs was less than observed for humans at the highest proposed therapeutic dose.
`
`2.4.3 Genotoxicity in vitro and in vivo
`Genotoxicity studies in vitro (bacterial mutation assays in salmonella and E.coli and chromosomal
`aberrations in CHO cells in absence and presence of metabolic activation) and in vivo (rat
`
`Page 6/30
`
`PAR1021
`IPR of U.S. Patent No. 8,772,306
`Page 6 of 30
`
`

`
`micronucleus test) did not identify a cause for concern. Sodium oxybate may be considered as non-
`genotoxic.
`
`2.4.4 Carcinogenicity (with toxicokinetics)
`There is available data on rat and mouse with γ-butyrolactone (GBL which converts in GHB rapidly in
`the body) in two NTP (National Toxicology Program) studies and with sodium oxybate in rats
`(applicant-sponsored 2-year study). In the rat carcinogenicity study the active substance was
`administered as sodium oxybate (maximum dose 1000 mg/kg). In the National Toxicology Program
`mouse carcinogenicity study (CAS No. 96-48-0) the active substance was administered as GBL
`(maximum dose 525 mg/kg). Provided that no toxicokinetic data was included in the NTP studies,
`single dose and 14 day dose bridging studies were conducted by the applicant in both rat and mouse to
`estimate the exposure in these species following administration of sodium oxybate or γ-butyrolactone
`(GBL) in support of these carcinogenicity studies. The exposures in rat and mouse at the doses
`corresponding to the maximum doses in the respective carcinogenicity studies was compared to the
`exposures in humans at the maximum recommended dose of 9g, administered as two 4.5g doses 4
`hours apart (OMC-SXB-9). The animal/human exposure ratios for Cmax were 2.60 and 2.76 for mouse
`and rat, respectively. Similarly the animal/human exposure ratios for AUC were 1.21 and 1.64 for
`mouse and rat, respectively. It is not clear whether data from short-term administration (up to 14 days)
`is informative regarding the exposure of animals at the end of the study, provided that accumulation
`seemed to occur at least at high doses, as suggested by comparative toxicokinetic analysis of the
`values obtained in repeated dose studies in rats and dogs. Underestimation of exposure may therefore
`be given in the bridging studies.
`GBL has been classified by NTP as non-carcinogenic in rats and equivocal carcinogen in mice, due to
`slight increase of pheochromocytomas which was difficult to interpret due to high mortality in the
`high dose group. With GBL a non-significant increase of hyperplasia of adrenal medulla was observed
`also in rats, which poses the possibility of a drug-related effect in both species. Decreased incidence of
`several neoplasm types in mice (hepatocellular tumors) and rats (mammary fibroadenomas)
`administered with GBL were also observed in the NTP study.
`In rats, both sodium oxybate (applicant-sponsored study) and GBL were classified as non-
`carcinogenic. In the oxybate study 2/50 pituitary carcinomas were observed in high-dose female rats
`compared to 0/50 in all other groups including controls. However, this finding was of doubtful
`statistical significance and the incidence was at the upper bound of historical controls. Since GHB was
`non-genotoxic and the pituitary carcinomas in the rat were of marginal statistical significance, there is
`sufficient information to assume that Xyrem is unlikely to be a potential carcinogen in humans. The
`safety ratios calculated against predicted human exposure were still low but new studies do not seem
`necessary as no concern has been raised from the available toxicological data.
`
`2.4.5 Reproductive and developmental studies
`GHB had no effect on mating, general fertility or sperm parameters and did not produce embryo-foetal
`toxicity in rats exposed to up 1000 mg/kg/day GHB. As there are no PK data in pregnant animals, the
`corresponding exposure margin, calculated from non-pregnant animals is 1.64 times the human one.
`In rabbits, foetotoxicity was slight and did not reach statistical significance. The only notable
`abnormality was hydrocephalus in two foetuses from the same litter in a mid-dose female. Based on
`historical data provided and taking into consideration that there were no similar findings in any other
`litter or dose group, this finding was considered to be unrelated to treatment. The rabbit study included
`toxicokinetics. The highest Cmax recorded was 454 µg/ml, which is 3-fold higher than the predicted
`human value.
`GHB had no adverse effects on the F0 animals in a conventional Segment III study in rats, except for
`an increase in the incidence of post-dose sedation, low bodyweight gain and reduced food
`consumption in the high-dose group. Perinatal mortality was increased and mean pup weight was
`decreased during the lactation period in high-dose F1 animals. Though not dose related, a relationship
`of these mortalities with the treatment cannot be ruled out. GHB is not to be recommended during
`pregnancy or breast-feeding and this is reflected in the SPC.
`
`Page 7/30
`
`PAR1021
`IPR of U.S. Patent No. 8,772,306
`Page 7 of 30
`
`

`
`2.4.6 Other toxicity studies
`Immunotoxicity studies were not carried out. However, there were no signals of direct immunotoxicity
`in any of the repeat-dose toxicity studies.
`Dependance studies
`GHB is a known substance of abuse and in the United States the use of Xyrem is subject to a rigid risk
`management program. The applicant did not conduct specific animal tests for dependence. However, a
`review of the published literature was carried out to collect information on the effects of GHB in
`models of drug discrimination, self-administration and tolerance. Drug discrimination studies show
`that GHB produces a unique discriminative stimulus that in some respects is similar to that of alcohol,
`morphine and certain GABA-mimetic drugs. However, the characteristics of these effects differ with
`dose, suggesting the involvement of multiple receptor systems with varied affinities for GHB. Self-
`administration studies in rats, mice and monkeys have produced conflicting results, whereas tolerance
`to GHB as well as cross-tolerance to alcohol has been clearly demonstrated in rodents.
`The data generated in the file does not allow evaluation of the potential of sodium oxybate to induce
`withdrawal phenomena. The literature review suggest that mild effects could occur in mice and rats
`after frequent daily administration and in primates after continuous long term administration (more
`than 30 days) of 750mg/kg/day dose. The potential for withdrawal seems therefore to exist, though
`limited. This is reflected in the SPC and will be further evaluated clinically as a post-marketing
`commitment.
`2.5 Environmental risk assessment
`A formal environmental risk assessment has not been carried out. It is nevertheless agreed that sodium
`oxybate is unlikely to pose any perceivable risk to the environment.
`
`2.6 Discussion on the non-clinical aspects
`The precise mechanism by which sodium oxybate produces an effect on cataplexy is unknown,
`however sodium oxybate is thought to act by promoting slow (delta) wave sleep and consolidating
`night-time sleep. The cause of human narcolepsy and cataplexy is, as yet, unknown. Recent evidence
`points to the loss of hypocretin-containing neurons, possibly due to autoimmune attack, as a likely
`cause. Animal models of cataplexy and narcolepsy are continuing to be developed, but the effects of
`GHB in these models, have yet to be investigated.
`In addition to its sedative properties, GHB is a potent CNS depressant and may increase growth
`hormone secretion. The potential for acute respiratory depression after reintroduction of treatment in
`patients who might have developed tolerance is mentioned in the SPC (4.2) as well as a warning in
`case of concomitant administration with other CNS depressant, and especially alcohol. (SPC, 4.4 and
`4.5)
`The interactions resulting from the stimulation or inhibition of GHB dehydrogenase, namely with
`anticonvulsivant drugs and L-dopa are considered to be potentially relevant in the clinic and are
`mentioned in the SPC.(section 4.5 and 5.3).
`The bioavailability in rats is about 50-80% and the clearance is rapid (T½ ≈1h) with extensive
`biotransformation which is not P450 dependant. The kinetic is non-linear, due to saturable
`mechanisms of both absorption and elimination resulting in reduced Cmax and increased AUC from
`linearity with increasing doses. As GHB is metabolised by γ hydroxybutyrate (GHB) dehydrogenase,
`there is a potential interaction with drugs that inhibit this enzyme. This is reflected in the SPC
`(sections 4.5 and 5.3)
`In repeat-dose toxicity, treatment-related clinical signs were mainly related to sedation. Safety
`margins, based on body weight changes and salivary gland atrophy, are low or non-existent. This is in
`accordance with the frequent occurrence in humans of adverse effects such as nausea, anorexia and
`parasympathetic disorders (blurred vision, enuresis and sweating).
`GHB is non-genotoxic and not considered to present a carcinogenic risk to humans.
`In reproductive toxicity studies, foetoxicity was observed in rats and rabbits. The use of GHB is not
`recommended during pregnancy or breast-feeding and this is reflected in the SPC.
`
`Page 8/30
`
`PAR1021
`IPR of U.S. Patent No. 8,772,306
`Page 8 of 30
`
`

`
`Tolerance to GHB as well as cross-tolerance to alcohol has been clearly demonstrated in rodents. The
`potential of GHB to induce withdrawal phenomena, demonstrated in animal models, is low, and the
`relevance for humans will be monitored i

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket