throbber
SCIENTIFIC DISCUSSION
`
`This module reflects the initial scientific discussion for the approval of Ammonpas. This
`scientific discussion has been updated untill November 2001. For information on changes after
`this date please refer to module 8B.
`
`1.
`
`Introduction
`
`Ammonaps, sodium phenylbutyrate (PB), is a new active substance. It is indicated as adjunctive
`involving deficiencies of
`the chronic management of urea cycle disorders,
`therapy
`in
`carbamylphosphate synthetase, ornithine transcarbamylase, or argininosuccinate synthetase. It is
`indicated in all patients with neonatal-onset presentation (complete enzyme deficiencies, presenting
`within the first 28 days of life). It is also indicated in patients with late-onset disease (partial enzyme
`deficiencies, presenting after the first month of life) who have a history of hyperammonaemic
`encephalopathy.
`
`The dossier submitted in support of the application comprises data generated by the applicant: all
`chemical/pharmaceutical data, the two mutagenicity studies for Part III, and for Part IV, the
`bioequivalence study and a review of the US IND/NDA programme. Additional information was
`available from published literature.
`
`Urea cycle disorders (UCD) are inherited deficiencies of one of the enzymes involved in the urea
`cycle, by which ammonium is converted to urea. Ammonium is highly toxic to nerve cells and
`hyperammonaemia may result in metabolic derangement, leading to anorexia, lethargy, confusion,
`coma, brain damage, and death.
`
`The most severe forms of UCDs occur early in life (complete enzyme deficiencies). The classic
`neonatal presentation of all the UCD (with the exception of arginase deficiency) is quite uniform and
`includes, after a short symptom-free interval of one to five days, poor feeding, vomiting, lethargy ,
`muscular hypotonia, hyperventilation, irritability and convulsions. Without rapid intervention, coma
`prevails as the condition worsens and leads eventually to deaths. Later onset forms of UCD occur in
`infancy, at puberty, and in adults subject to physiological stress. In the late onset forms, more subtle
`symptoms have been described including vomiting, migraine-like headache, changes in the level of
`consciousness and neurological signs, such as
`lethargy, somnolence,
`irritability, agitation,
`combativeness, disorientation, ataxia and visual impairment. Seizures are a late complication. Finally,
`delayed physical growth and delay in mental development are common. In female patients with
`ornithine transcarbamy lase deficiency, who are heterozygous, the condition is less severe and they
`may remain undiagnosed well into adult life.
`
`In the absence of systematic screening, the incidence of UCD is difficult to assess and various
`estimates are found in the literature. On this basis, it is estimated that the overall incidence of all urea
`cycle disorders has been defined as 1 per 8,200 births.
`
`The treatment strategies used are to reduce dietary protein intake, and to provide an alternative vehicle
`to urea for the excretion of nitrogen waste. Currently none of the possible treatments for
`hyperammonaemia are approved in Europe. Enzyme replacement therapy through liver transplantation
`provides an additional treatment option. In most patients this procedure has markedly improved their
`metabolic abnormalities and permitted a normal protein intake, however, transplantation for UCD is a
`relatively recent treatment option and its long-term benefits are as yet unknown.
`
`Sodium phenylbutyrate is a prodrug and is rapidly metabolised to phenylacetate. It promotes the
`synthesis of phenylacetylglutamine, which then serves as a substitute vehicle for waste nitrogen
`excretion. The recommended dose is:
`
`450 - 600 mg/kg/day in neonates, infants and children weighing less than 20 kg
`9.9- 13.0 g/m2/day in children weighing more than 20 kg, adolescents and adults.
`The safety and efficacy of doses in excess of 20 g/day has not been established.
`
`1/12
`
`© EMEA2005
`
`Par Pharmaceutical, Inc. Ex. 1019
`Par v. Horizon, IPR of Patent No. 9,561,197
`Page 1 of 12
`
`

`

`2.
`
`Chemical, pharmaceutical and biological aspects
`
`Composition
`
`Ammonaps is presented as tablets and granules containing sodium phenylbutyrate. Two standard and
`simple pharmaceutical formulations of sodium pheny lbutyrate were produced with commonly used
`excipients. The tablets (500 mg) contain approximately 74% active substance and the granules provide
`940 mg sodium phenylbutyrate/g granules.
`
`In order to dose accurately and especially for smaller amounts required for infants, three measuring
`spoons have been introduced for the granules, giving doses of 0.95 g, 2.9 g and 8.6 g. The overall
`uniformity of doses obtained from the three measuring spoons is acceptable and the individual weights
`are within the Ph. Eur. limit of 10% for single-dose powders.
`
`The proposed container for both granules and tablets is a high density polyethylene (HDPE) bottle
`with a desiccant unit, closed with a polypropylene caps (child resistant). The materials have been
`adequately tested for conformance to USP requirements.
`Active substance
`
`Pharmaceutical data on the active substance have been presented in an EDMF (European Drug Master
`File). Sodium phenylbutyrate is off-white to slightly yellow powder, which is soluble in water. A four(cid:173)
`step synthetic process with acceptable in-process controls manufactures it. Process validation data
`show the synthesis to be under control. Satisfactory specifications were provided for the starting
`material, solvents, reagents and intermediates. The manufacturer of the active substance has
`adequately validated the analytical methods used. The manufacturer of the finished product to re-test
`the active substance uses the same methods; full re-validation is to be carried out on these methods
`and results provided.
`
`Sodium phenylbutyrate has a simple structure and presents no polymorphic forms. The pathway of
`synthesis has confirmed the evidence of its chemical structure, by elemental analysis, 1 RNMR and IR
`spectroscopy.
`
`The specification includes tests for appearance, bulk density, water content, identifcation by IR and
`HPLC, heavy metals, pH, assay and impurities. Three main related substances are specified: a(cid:173)
`tetralone, 3-benzoylpropionic acids and 4-cyclohexylbutyric acids. Further impurities (e.g. isomer 2-
`phenylbutyric acid) can be detected by a GC or HPLC assay method but have not been found in the
`active substance. While the limits for impurities have been toxicologically accepted, it is suggested
`that in view of the high doses to be given (> 2 g/day), limits should be reviewed and tightened when
`further batch data are available. Residual solvents are also specified at a suitable limit in agreement
`with CPMP/ICH guidance.
`
`Analytical results from three batches show compliance with the specification and indicate suitable
`uniformity.
`
`The active substance (3 batches) was tested for up to 12 months under real-time (25°C/60%RH) and
`accelerated condition ( 40°C/75%RH). It was also tested in solvent and solution, under the influence of
`pH and oxidative conditions. The shelf-life specification includes appearance, assay, impurities, pH
`and water. Increases in water content were observed but are not linked to the increases in 3-
`benzoy I propionic acid also seen in stability batches and are not detrimental to the stability of sodium
`phenylbutyrate. A 12-month retest period can be approved.
`Other ingredients
`
`Satisfactory information has been provided on the excipients. All excipients will be released against
`relevant Ph.Eur. Monographs. For those excipients derived from tallow (i.e. magnesium and calcium
`stearate), a TSE declaration was provided in accordance with the EU requirements (Commission
`Decision 97/534/EC).
`
`2/12
`
`©EMEA2005
`
`Par Pharmaceutical, Inc. Ex. 1019
`Par v. Horizon, IPR of Patent No. 9,561,197
`Page 2 of 12
`
`

`

`Product development and finished product
`
`No detailed pre-formulation studies were performed. The tablets and granules are manufactured using
`simple formulations based on commonly used excipients, standard pharmaceutical equipment and
`processes. The function of the excipients is stated.
`
`Forced degradation studies have been conducted under extreme temperature and acidic conditions.
`They indicate a rise in 3-benzoylpropionic acid level, as well as some degradants not detectable by
`HPLC, but these extreme conditions do not reflect the product as marketed. Results of up to 0.006%
`w/w were found from batches tested for 3-benzoylpropionic acids.
`
`Batches manufactured at different sites have been used in clinical trials and bioequivalence studies.
`Results of a three-way crossover study in healthy volunteers receiving 5 grams doses of tablets or
`granules indicate that the bioavailability of the granule formulation is less than that of the tablets, but
`remains within the usual criterion of± 20%. This will be further discussed in Part IV.
`
`The manufacturing processes for both granules and tablets consist of multi-stage blending,
`compaction, granulation, and compression as the final step for tablets. The processes are satisfactorily
`described.
`
`Mixing times, equipment conditions and in-process controls are described for both formulations
`accordingly (weight, thickness, hardness, friability for the tablets, fill volume for the granules and bulk
`and tapped density testing for both tablet and the granules) and their parameters are specified within
`acceptable limits. Results from clinical (7 and 5 batches for granules and tablets, respectively) and
`production (2 batches for granules and tablets) batches indicate acceptable batch-to-batch consistency.
`
`A revised finished product specification (for both the site of manufacture and the site of batch release)
`has been provided in compliance with EU requirements. Control tests on the finished product use
`adequately validated methods and include requirements for appearance, identification of active
`substance, assay and impurities determination, bulk density testing for granules, and average weight,
`uniformity of weight, disintegration and dissolution for the tablets. The microbiological quality is
`controlled in accordance with Ph. Eur., but is proposed as a non-routine method.
`
`The dissolution medium, previously simulated intestinal fluid, has been changed to water. The
`dissolution specification has been tightened to 80% in 45 minutes but this should be reviewed again in
`the light of further data. Dissolution results using both media show slightly greater dissolution in
`water, but dissolution is essentially complete in both media at the same time. The disintegration limit
`is set slightly higher than usual (at 20 min); this is acceptable as the results do not impact adversely on
`dissolution.
`
`A commitment is given by the applicant to submit certificates of analysis for the first three production
`batches, tested to the EU specifications. Limits for impurities will be reviewed when further batch data
`are available.
`
`On the basis of the inspection carried out at Pharmaceutics International Inc on 13-15 May 1998, the
`inspection report confirmed that the operations are in general compliance with the principles and
`guidelines of GMP (see the Annex II).
`
`Stability studies have been carried out at 25°C/60%RH up to 24 and 36 months on batches of granules
`and tablets made by Pharmaceutics International, and at 40°C/75%RH for 6 months. Shelf-life content
`limits of 93-107% have been accepted for the finished products on the basis of the variability in
`results, though no degradation appears to occur. The limits should be reviewed again when further
`stability data are available. The analytical methods used are those for routine finished product testing
`or similar, validated methods. No change of appearance was observed. Content and impurity levels
`remain within the proposed limits as specified. Satisfactory stability data for the full shelf-life have
`been provided and based on the resulting data, a 2-year shelf life is acceptable for both granules and
`tablets when stored below 30°C.
`
`•
`
`Discussion on chemical, pharmaceutical and biological aspects
`
`Ammonaps granules and tablets are conventionally formulated and manufactured using standard
`pharmaceutical technology. A suitable specification has been submitted for the active substance. The
`limits for impurities have been toxicologically accepted (see Part III). A single specification for each
`
`3/12
`
`©EMEA2005
`
`Par Pharmaceutical, Inc. Ex. 1019
`Par v. Horizon, IPR of Patent No. 9,561,197
`Page 3 of 12
`
`

`

`finished product formulation is also proposed, with revised specification for dissolution parameters
`and impurity limits. In line with the requirements for the active substance, the impurity limits should
`be reviewed when further data are available.
`
`Overall, the chemical-pharmaceutical dossier is generally acceptable. The company was however
`requested to provided, within the agreed timeframe, additional data, which have not been satisfactorily
`resolved; these are defined in the follow-up measures as listed in the company's undertaking letter (see
`section II.3 of this report).
`
`3.
`
`Toxico-pharmacological aspects
`
`Pharmacodynamics
`
`Pharmacodynamic effects relating to the proposed indications are as outlined in section 4 (Clinical
`pharmacology /Pharmacodynamics).
`
`General pharmacodynamics - A number of studies seem to indicate the ability of PB to inhibit tumour
`growth in vitro, and that phenylacetate and probably phenylbutyrate have neuroinhibitory and
`neurotoxic potential under the in vitro and ex-vivo conditions studied.
`
`Two rat models of human phenylketonuria were developed, one involved exposure to PA injected s.c.
`twice daily from day 2-28 of life. In the other, pregnant rats were exposed to P A during gestation.
`Reduced brain weight, abnormalities in learning, and in neurotransmitter uptake are consistently
`noted. It was argued that high concentrations are unlikely during therapeutic use of PB because of
`poor transfer across the adult blood-brain barrier. The implications of these findings with respect to
`human foetal brain are unknown (see also below- Reproductive and development toxicity studies)
`
`Pharmacokinetics
`
`Studies in the juvenile rat, where subcutaneous administration was used, and in the adult cat, where
`intravenous administration was used, have been performed. Even though pharmacokinetic data after
`oral administration are not available, it can be expected that being an organic acid, PB will be rapidly
`and extensively absorbed after oral administration. It is converted to its active metabolite, P A by beta(cid:173)
`oxidation. In single subcutaneous dose studies from birth to maturity in rats, P A penetrated tissues
`rapidly and extensively, with tissue levels usually equivalent to those in blood. Like other organic
`acids, P A is actively excreted in urine by tubular secretion as the amino acid conjugate.
`Toxicology
`Single dose toxicity - No single dose toxicity studies have been carried out. However, sufficient
`information is available from the animal pharmacology above. The doses of P A given in these studies
`were low. Taken together, the results of the studies suggest that single doses of PA by both the
`intravenous and subcutaneous routes are well tolerated.
`
`Repeated-dose toxicity - There are no repeated dose studies available. However, information available
`from the animal pharmacology above makes a convincing case that parenteral administration of
`phenylacetate causes impairment of brain development in the immature rodent. Because phenylacetate
`can cross into human CNS, the observations in rodents should be considered a potential hazard for the
`therapeutic use of PB.
`
`Carcinogenicity - Carcinogenicity studies have not been performed. These deficiencies are not
`considered to be an impediment to the granting of a Marketing Authorisation in view of ICH-S1A:
`guideline on the need for carcinogenicity studies of pharmaceuticals.
`
`Genotoxicity and mutagenicity - A bacterial reverse mutation assay (Ames test, plate incorporation
`method) was conducted with PB at concentrations in the range of 52-5000 J.Lg/plate, using five strains
`of Salmonella Typhimurium, in the presence and absence of rat liver microsomal enzymes (S9). No
`cytotoxicity or revertant colonies were observed at the top dose. A bone marrow micronucleus test
`was also conducted using rats of both sexes (5 animals/sex/group; PB 878-1568-2800 mg/kg single
`oral gavage). Deaths occurred in top dose (7/10, at 2800 mg/kg) and mid-dose (2/10, at 1568 mg/kg)
`groups. The frequency of micronucleated cells was not significantly different from the negative
`control at any dose level at either the 24 hour or the 48 hour harvest.
`
`4/12
`
`© EMEA2005
`
`Par Pharmaceutical, Inc. Ex. 1019
`Par v. Horizon, IPR of Patent No. 9,561,197
`Page 4 of 12
`
`

`

`Attention should be drawn to the fact that the Ames test did not comply with the ICH-requirements
`(i.e. two recommended strains of E. coli were not included to pick-up A-T and G-C base pair
`mutations) and there are no pharmacokinetic data in either rat or man to validate the in vivo study in
`terms of reaching adequate plasma levels. Despite these deficiencies, the results of both studies did not
`give raise to any evidence of mutagenic potential.
`
`Reproductive and development toxicity studies - Studies on administration to pregnant rodents indicate
`that CNS damage may occur in animals exposed in utero. However, as drug administration did not
`commence until day 9 of gestation, after the main period of organogenesis, these studies are not
`optimal for the assessment of teratogenic potential. In female pregnant rats, spontaneous abortions
`occurred, birth weight of the offspring was significantly lower than in controls, weight gain of the
`pups over the lactation period was reduced, and brain weight at sacrifice was low. It also seems likely
`that spermatogenesis and therefore fertility would be affected in the male rat.
`
`Impurities- In the active substance, a-tetralone, 3-benzoylpropionic acid and 4-cyclohexylbutyric acid
`are the potential impurities identified. According to the ICH requirements, the threshold for
`toxicological qualification of impurities is 0.05% (w/w) and of degradation products is 0.1 %, when the
`total daily intake exceeds 2 g, as in the case of Ammonaps. The limits for cyclohexylbutyric acid and
`for other impurities in the active substance and in the release specification for the tablets and granules
`are higher than the threshold (at 0.1 %), but the limits have been found to be toxicologically
`acceptable. No adverse events would be expected as a result of these impurities, but the applicant is
`required to submit further data from manufacturing batches and these data will be reviewed (see also
`Part II).
`
`•
`
`Summary and conclusion on preclinical pharmacology and toxicology:
`
`There are no formal toxicity studies; no overt toxicity was noted in a review of the data available. A
`bacterial reverse mutation and a rat bone marrow micronucleus test have been carried out with sodium
`phenylbutyrate and did not give rise to any evidence of mutagenic potential. The available data
`indicate that PB is fetotoxic, affecting mainly the brain; effects on reproduction and organogenesis
`have not been conventionally investigated. This has been dealt with in the SPC, where pregnancy is
`contra-indicated and an explanation is given in the appropriate section of the document.
`
`The deficiencies of the pre-clinical section of the dossier should be viewed in the light of the CPMP
`recommendation for an approval under exceptional circumstances. As required for an authorisation
`under exceptional circumstances, appropriate information is provided in the product information to
`draw the attention of the medical practitioner to the fact that the currently available data concerning
`the medicinal product in question is inadequate in certain specified respects. The conditions for which
`this medicinal product would be indicated would fall within the scope of the Proposed European
`Parliament and Council Regulation (EC) on Orphan Medicinal Products.
`
`4.
`
`Clinical aspects
`
`Ammonaps, sodium phenylbutyrate (PB), is a new active substance with the proposed therapeutic
`indication "adjunctive therapy in the chronic management of urea cycle disorders, involving
`deficiencies of carbamy 1 phosphate synthetase, ornithine transcarbamy lase or argininosuccinate
`synthetase". Urea Cycle Disorders (UCD) are inherited deficiencies of one of the enzymes involved in
`the urea cycle, by which ammonium is converted to urea. Excess dietary protein and the nitrogenous
`substances produced by endogenous protein turnover are normally metabolised to yield energy and the
`by-product ammonium, which is excreted in the urine as urea. Each pass through this cycle results in
`the elimination of one molecule of urea, which contains two atoms of waste nitrogen. Due to
`deficiencies of the urea cycle, the conversion of ammonium ion to urea is impaired to varying degrees,
`and consequently
`its excretion is reduced. Ammonium is highly toxic
`to nerve cells and
`hyperammonaemia can damage the central nervous system leading to cerebral oedema and death.
`
`The elimination of nitrogen from the human body by a moiety other than urea was first proposed in
`1914, when Lewis described the stoichiometric relationship between the decrease in urine nitrogen as
`urea and the appearance of hippurate nitrogen in a normal subject given sodium benzoate.
`Subsequently, Sherwin in 1919 demonstrated the quantitative elimination of nitrogen in humans via
`
`5/12
`
`©EMEA2005
`
`Par Pharmaceutical, Inc. Ex. 1019
`Par v. Horizon, IPR of Patent No. 9,561,197
`Page 5 of 12
`
`

`

`pheny lacety I glutamine following treatment with oral doses of phenylacetic acid (P A). The amino acid
`acylation products of sodium benzoate and sodium phenylacetate may substitute for urea nitrogen
`excretion in all UCD.
`
`The investigational use of P A for the treatment of patients with urea cycle disorders was started in
`clinical trials performed at Johns Hopkins University, USA, in 1980. Subsequently, the relevant
`permission was amended to include the investigational use of the combination of sodium benzoate and
`PA at several dosages. In 1983, a further amendment permitted the use of PB as a substitute for PA.
`Finally, in 1987, the use of PB only was introduced as a monotherapy replacing the combination
`therapy.
`
`The Office of Orphan Products Development supported the trial conducted with PB and the US
`Orphan status designation was granted on 22 November 1993. The results of this trial constitute the
`basis of the clinical part of the dossier. Although this trial does not comply with the requirements of
`Good Clinical Practice, it seems that the study population represents a significant proportion of
`patients with these rare disorders treated in the USA.
`Clinical pharmacology
`
`Pharmacodynamics - PB is a pro-drug, which is rapidly converted to P A by beta-oxidation in
`mammalian liver and kidneys. In higher primates, P A is enzymatically conjugated with glutamine in
`the liver and kidneys to form pheny lacety !glutamine (P AG), which is readily excreted in the urine.
`The glutamine required to excrete the P A will have to be synthesised and the reversible reaction:
`glutamine Bglutamic acid + NH4+ will proceed to the left and therefore ammonia will be excreted
`This cannot be demonstrated in animals due to species differences in the metabolic pathway of
`nitrogen elimination. On a molar basis, P AG is comparable to urea (each containing two nitrogen
`atoms) and provides an alternative vehicle for waste nitrogen disposal. Thus therapeutic administration
`of PB has the potential to divert nitrogen away from the blocked or impaired urea cycle and to provide
`an alternative pathway of excretion.
`
`Experimental support for the hypothesis outlined above is provided by the work of Prof. Brusilow's
`group at the Johns Hopkins school of Medicine. Brusilow demonstrated in 1991 in a child with
`carbamy 1 phosphate synthetase deficiency that administration of P A or PB resulted in the urinary
`excretion of P AG equivalent to 38-44% of the predicted normal nitrogen excretion. In other children
`with UCD, administration of PB or PA resulted in a decrease of 25-50% of baseline glutamine.
`Similar results were found in an adult male patient with ornithine transcarbamy lase deficiency, whose
`plasma ammonium and glutamine levels significantly declined with PB therapy.
`
`Pharmacokinetics - No formal pharmacokinetic studies have been performed with PB. Data from
`bioequivalence studies, and pilot studies in patients with cancer and haemoglobinopathies are cited.
`
`Studies in cancer patients have been performed where intravenous infusion of PB or P A has been used
`infusion, PB and PA display non-linear
`for anti-tumour activity. After
`intravenous bolus
`pharmacokinetics with a saturable elimination, which is consistent with an enzymatic process. During
`treatment with repeated doses of pheny lacetate there is evidence of an induction of drug clearance as
`shown by a significant decrease (27%) in the AUCs obtained at the beginning (days 1-3) compared
`with those in the end (days 12-14) of therapy, which the authors attribute to enzyme induction.
`Concentrations over 900 Jlg/ml were associated with sedation, confusion, nausea and vomiting.
`
`Pharmacokinetics after oral administration of PB have been studied in healthy volunteers (single dose
`of 2.5 g, n=2; single dose of 5 g, n= 21), in one patient with ornithine transcarbamylase deficiency and
`in 8 patients with haemoglobinopathies. PB is rapidly absorbed: measurable plasma levels of PB are
`detected 15 min after oral administration. Peak concentrations of approximately 1 mmol/1 are reached
`after 1 h. In one study, the elimination half-life was estimated to be 0.8 h. Measurable plasma levels of
`PA and PAG are detected 30-60 min after oral dosing ofPB (the mean peak concentration is 45.3 and
`62.8 Jlg/ml, respectively). The time to peak concentration increases with the dose of PB and is around
`3.5 h for both metabolites after a dose of 5 g of PB. The elimination half-life was estimated to be 1.3
`and 2.4 hours, respectively for P A and P AG. Recovery of PB and P AG from serial collections of urine
`has been evaluated in some of the cited studies. It is demonstrated that in most subjects, the kidneys
`within 24h excrete approximately 80-100% of the drug as the conjugated product, P AG. After oral
`administration, unchanged drug is not detected in the urine of normal subjects or patients with UCD. It
`
`6/12
`
`© EMEA2005
`
`Par Pharmaceutical, Inc. Ex. 1019
`Par v. Horizon, IPR of Patent No. 9,561,197
`Page 6 of 12
`
`

`

`has not been determined if P A is secreted in human milk, therefore Ammonaps is contraindicated
`during breast-feeding.
`
`For all formulations used in the bioequivalence studies, the total exposure was greater in female than
`in male subjects, being about 20-25% and 40% higher for PB and PA, respectively. This gender
`difference has been mentioned in the SPC. This may be due to the lipophilicity of the drug and gender
`differences in volume of distribution. For PAG the difference was only about 10%. The principal
`indices of plasma pharmacokinetics of PB, P A and P AG in healthy male and female subjects are
`tabulated below (Table 1).
`
`Table 1 - Plasma pharmacokinetics (mean values) in healthy male (m) and female (f) subjects:
`
`tmax (h)
`M 1.18
`Pheny I butyrate
`F 1.21
`(PB)
`M3.62
`Pheny lacetate
`(PA)
`F 3.73
`M3.25
`Pheny lacety 1
`glutamine (P AG) F 3.43
`
`Cmax (J.lg/ml)
`M 192.5
`F 242.6
`M39.2
`F 55.1
`M 67.4
`F 66.9
`
`AUC (J.lg·h/ml)
`M 480.1
`F 622.1
`M 154.4
`F 245.8
`M 282.7
`F 297.7
`
`tvz (h)
`M0.78
`F 0.82
`M 1.2
`F 1.26
`M2.12
`F 2.66
`
`Bioequivalence studies - In a single dose three-way crossover study, healthy male (n=lO) and female
`(n= ll) volunteers received a) PB 500 mg tablets, American Drug Development Inc., b) PB powder,
`Pharmaceutical services University of Iowa or c) PB 500 mg tablets, Pharmaceutical Services
`University of Iowa. In all cases the dose was 5 grams. A and B were considered the test and C the
`reference formulations. Results are tabulated below (Table 2) and indicate that the bioavailability of
`the powder formulation is less than that of the tablets, but remains within the usual regulatory criterion
`of ± 20%.
`
`Table 2 - Relative bioavailability (mean values) of test tablets (a) and powder (b) compared to
`reference tablets (c):
`
`Pheny I butyrate
`(PB)
`
`tmax (h)
`a) 1.4
`b) 1.0
`c) 1.2
`d) 3.7
`e) 3.6
`f) 3.7
`g) 3.4
`Pheny lacety 1
`glutamine (P AG) h) 3.2
`i) 3.4
`
`Pheny lacetate
`(PA)
`
`Cmax (J.lg/ml)
`a) 218
`b) 195
`c) 240
`d) 49
`e) 45
`f) 54
`g) 69
`h) 63
`i) 69
`
`AUC (J.lg·h/ml)
`a) 577
`b) 494
`c) 586
`d) 211
`e) 188
`f) 231
`g) 306
`h) 268
`i) 301
`
`tvz (h)
`a) 0.77
`b) 0.76
`c) 0.85
`d) 1.15
`e) 1.29
`f) 1.25
`g) 2.41
`h) 2.36
`i) 2.56
`
`All formulations in the bioequivalence study contained approximately the same amount of active
`substance and relatively minor differences in excipients. No safety problems have been observed from
`clinical experience with the proposed commercial formulation, as distinct from development
`formulations .
`
`Interaction studies -No studies of drug interactions are included in support of the application. There
`have been publications reporting that: renal excretion of the PB conjugation product may be affected
`by concurrent administration of probenecid; hyperammonia may be induced by haloperidol and by
`valproate; corticosteroid may cause increase in plasma ammmonia levels. More frequent monitoring of
`plasma ammonia levels is advised in the SPC when using these medications.
`
`There is no formal food interaction study available. The suggestion for administration with food is
`based on experience gained in clinical practice. In addition, the dose is to be titrated against metabolic
`states and an important principle would seem to be that the administration with or without is kept
`constant.
`
`Special patient groups - The pharmacokinetics of PB have been studied in male patients with hepatic
`cirrhosis (oral administration, 20 g/day in three doses). Plasma levels of PB followed the peak and
`
`7/12
`
`©EMEA2005
`
`Par Pharmaceutical, Inc. Ex. 1019
`Par v. Horizon, IPR of Patent No. 9,561,197
`Page 7 of 12
`
`

`

`trough pattern familiar from healthy subjects and UCD patients. The conversion of P A to P AG was
`relatively slower in these patients with impaired hepatic function as evidenced by progressive
`accumulation of P A in the plasma of 3 out of the 6 patients studied; this pattern is not found in
`subjects with normal liver function. In addition, PB and P A were detected in urine. This suggests that
`in patients with cirrhosis the capacity of the metabolic pathway of P A is reduced.
`
`The pharmacokinetics of PB have not been studied in patients with renal impairment.
`
`Appropriate information concerning these high risk groups is contained in the SPC.
`
`Dosage -No formal dose finding study has been performed. The proposed daily dosage was derived
`on the basis that one mole of PB will be metabolised to one mole of P AG, and from the estimated
`nitrogen to be excreted on a restricted intake. Excretion of 0.09 g/kg/d of PAG nitrogen would require
`a dose of 0.6 g/kg/d of PB. It is likely that the efficiency of excretion varies between patients and
`individual titration is recommended on the basis of therapeutic monitoring. Based on this reasoning,
`the following regimen is proposed:
`
`450 - 600 mg/kg/day in neonates, infants and children weighing less than 20 kg
`9.9- 13.0 g/m2/day in children weighing more than 20 kg, adolescents and adults.
`
`Clinical efficacy
`
`The most important efficacy data are derived from the Phase III clinical trial, based on the US(cid:173)
`IND/NDA program. In this study, patients with UCDs [deficiency of carbamyl phosphate synthetase
`(CPS), ornithine transcarbamylase (OTC) or argininosuccinate synthetase (ASS)] were enrolled in an
`open, non-comparative multicentre study. The first patient was enrolled in 1985 and the cut-off for
`data analysis was February 1996. The IND program consisted of two cohorts of data, as outlined
`below.
`
`Study population - The first treatment program (1985-1994) consists of 162 patients, of which 148
`were evaluable (87 with prior therapy and 61 without prior therapy). The following UCDs had been
`diagn

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