throbber
Molecular Genetics and Metabolism 103 (2011) 323–329
`
`Contents lists available at ScienceDirect
`
`Molecular Genetics and Metabolism
`
`j ou r n a l h o m e pa g e : ww w. e l s ev i e r. c o m/ l o c a t e / y mg m e
`
`Ammonia control in children with urea cycle disorders (UCDs); Phase 2 comparison
`of sodium phenylbutyrate and glycerol phenylbutyrate☆
`Uta Lichter-Konecki a,⁎, G.A. Diaz b, J.L. Merritt II c, A. Feigenbaum d, C. Jomphe e, J.F. Marier e, M. Beliveau e,
`J. Mauney f, K. Dickinson g, A. Martinez g, M. Mokhtarani g, B. Scharschmidt g, W. Rhead h
`a Children's National Medical Center, 111 Michigan Ave NW, Washington, DC, 20010, USA
`b Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY, USA
`c Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA, USA
`d The Hospital for Sick Children, 555 University Avenue, Toronto Ontario, Canada
`e Pharsight, Montreal, 2000 Peel St., Suite 570, Quebec, Canada
`f Chiltern, 2520 Independence Blvd., Ste. 202, Wilmington NC, USA
`g Hyperion Therapeutics, Inc., 601 Gateway Blvd., Ste. 200, South San Francisco, CA, USA
`h Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI, USA
`
`a r t i c l e
`
`i n f o
`
`a b s t r a c t
`
`Article history:
`Received 17 February 2011
`Received in revised form 21 April 2011
`Accepted 21 April 2011
`Available online 5 May 2011
`
`Keywords:
`Urea cycle disorders
`Ammonia scavengers
`Hyperammonemia
`Phenylacetate
`Sodium phenylbutyrate
`Glycerol phenylbutyrate
`
`Twenty four hour ammonia profiles and correlates of drug effect were examined in a phase 2 comparison of
`sodium phenylbutyrate (NaPBA) and glycerol phenylbutyrate (GPB or HPN-100), an investigational drug
`being developed for urea cycle disorders (UCDs).
`Study Design: Protocol HPN-100-005 involved open label fixed-sequence switch-over from the prescribed
`NaPBA dose to a PBA-equimolar GPB dose with controlled diet. After 7 days on NaPBA or GPB, subjects
`underwent 24-hour blood sampling for ammonia and drug metabolite levels as well as measurement of
`24-hour urinary phenyacetylglutamine (PAGN). Adverse events (AEs), safety labs and triplicate ECGs
`were monitored.
`Results: Eleven subjects (9 OTC, 1 ASS, 1 ASL) enrolled and completed the switch-over from NaPBA (mean
`dose=12.4 g/d or 322 mg/kg/d; range=198–476 mg/kg/d) to GPB (mean dose=10.8 mL or 0.284 mL/kg/d
`or 313 mg/kg/d; range = 192–449 mg/kg/d). Possibly-related AEs were reported in 2 subjects on NaPBA
`and 4 subjects on GPB. All were mild, except for one moderate AE of vomiting on GPB related to an
`intercurrent illness. No clinically significant laboratory or ECG changes were observed. Ammonia was
`lowest after overnight fast, peaked postprandially in the afternoon to early evening and varied widely over
`24 h with occasional valuesN 100 μmol/L without symptoms. Ammonia values were ~25% lower on GPB vs.
`NaPBA (p≥ 0.1 for ITT and p b 0.05 for per protocol population). The upper 95% confidence interval for the
`difference between ammonia on GPB vs. NaPBA in the ITT population (95% CI 0.575, 1.061; p = 0.102) was
`less than the predefined non-inferiority margin of 1.25 and less than 1.0 in the pre-defined per-protocol
`population (95% CI 0.516, 0.958; p b0.05). No statistically significant differences were observed in plasma
`phenylacetic acid and PAGN exposure during dosing with GPB vs. NaPBA, and the percentage of orally
`administered PBA excreted as PAGN (66% for GPB vs. 69% for NaPBA) was very similar. GPB and NaPBA dose
`correlated best with urinary-PAGN.
`Conclusions: These findings suggest that GPB is at least equivalent to NaPBA in terms of ammonia control,
`has potential utility in pediatric UCD patients and that U-PAGN is a clinically useful biomarker for dose
`selection and monitoring.
`
`© 2011 Elsevier Inc. All rights reserved.
`
`Abbreviations: ASL, argininosuccinate lyase deficiency; ASS, argininosuccinate synthetase deficiency; AUC0–24, 24 h area under the curve; CV%, coefficient of variation; DSMB,
`Data Safety and Monitoring Board; GPB, glycerol phenylbutyrate (generic name for glyceryl tri (4-phenylbutyrate), also referred to as HPN-100); ITT, intention to treat; NaPBA,
`sodium phenylbutyrate; NH324-hour AUC, ammonia 24-hour area under the curve; OTC, ornithine transcarbamylase deficiency; PAA, phenylacetic acid; PAGN, phenylacetylglutamine;
`PBA, phenylbutyric acid; PK, pharmacokinetic; UCD, urea cycle disorder; ULN, upper limit of normal; U-PAGN24-hour Excr, PAGN excreted in urine over 24 h.
`☆ ClinicalTrials.gov identifier: NCT00947544.
`⁎ Corresponding author at: Department of Pediatrics, George Washington Univ. Medical Center, Division of Genetics and Metabolism, Children's National Medical Center,
`111 Michigan Ave NW, Washington, DC 20010, USA.
`E-mail address: ulichter@cnmc.org (U. Lichter-Konecki).
`
`1096-7192/$ – see front matter © 2011 Elsevier Inc. All rights reserved.
`doi:10.1016/j.ymgme.2011.04.013
`
`
`Page 1 of 7
`
`Owner Ex. 2018
`Par Pharm. v. Horizon
`IPR2015-01117,-01127
`
`

`
`324
`
`1. Introduction
`
`U. Lichter-Konecki et al. / Molecular Genetics and Metabolism 103 (2011) 323–329
`
`Urea cycle disorders (UCDs), which comprise several inherited
`enzyme and transporter deficiencies, result in the accumulation of toxic
`levels of ammonia in the blood and brain and can present in the neonatal
`period or later in life depending on the severity and type of defect [1–3].
`Control of hyperammonemia, the major cause of morbidity and
`mortality in UCD patients, is a major objective of treatment [4,5].
`Batshaw, Brusilow and coworkers introduced the concept of
`alternative pathway pharmacological therapy for UCDs [4,6,7]. These
`pioneering studies led to the availability of sodium phenylbutyrate
`(NaPBA), which is approved in the US (trade name: BUPHENYL®)
`(sodium phenylbutyrate) Powder and Tablets) and Europe (trade
`name: AMMONAPS®) for the chronic treatment of UCDs involving
`deficiencies of carbamylphosphate synthetase (CPS), ornithine trans-
`carbamylase (OTC), or argininosuccinic acid synthetase (AS) and
`lowers ammonia by enhancing excretion of waste nitrogen in the
`form of phenylacetylglutamine (PAGN). Despite the fact that
`alternative pathway therapy with NaPBA has been utilized for over
`two decades, there are little systematically collected data available
`pertaining to fasting and postprandial ammonia levels or correlates of
`drug effect in pediatric patients [8]. GPB is an investigational agent
`being developed for UCDs. Like NaPBA, it contains phenylbutyric acid
`(PBA), a pro-drug that is converted via β-oxidation to the active
`moiety, phenylacetic acid (PAA), which combines with glutamine to
`form PAGN that is excreted in the urine. However, unlike NaPBA
`which is a salt, GPB is a pre-pro-drug that contains no sodium
`and consists of three molecules of PBA joined to glycerol in ester
`linkage. It is hydrolyzed in the small intestine by pancreatic lipases to
`release PBA and glycerol, PBA is absorbed more slowly than when
`administered as NaPBA, and the glycerol is presumably digested
`like dietary glycerol consumed in the form of long chain triglycerides
`[8–10].
`
`2. Materials and methods
`
`2.1. Study design and treatments
`
`This phase 2, open-label, fixed sequence, switch-over study enrolled
`pediatric patients ages 6 or above being treated with NaPBA for a UCD
`(confirmed via enzymatic, biochemical or genetic testing). Major
`exclusion criteria included liver transplant, hypersensitivity to PBA,
`PAA or PAGN, clinically significant laboratory abnormalities or ECG
`findings, or conditions or medications that could affect ammonia levels.
`Enrolled subjects received NaPBA for at least 7 days, three times
`daily with meals at the dose level prescribed by the investigator. On the
`last day of NaPBA treatment they were admitted to a study unit and
`underwent 24-hour blood sampling for pharmacokinetic (PK) and
`ammonia measurements. Subjects were then switched to GPB at the
`PBA molar equivalent of their prescribed NaPBA dose. Initiation of GPB
`dosing was done under observation in an appropriately monitored
`setting and subjects were discharged after they were deemed clinically
`stable. After 7 days of treatment with GPB subjects were re-admitted to
`the research unit for 24-hour PK and ammonia monitoring, after which
`they were offered enrollment into a long-term extension study. Only the
`results of the switch-over part of the protocol are reported here.
`Subjects received dietary counseling and remained on their
`prescribed amount of dietary protein throughout the study. Diet
`was carefully monitored on study days 7 and 14 during the 24-hour
`blood sampling as well as for at least 3 days prior to each visit via
`diaries. Subjects also were queried at the end of the study with respect
`to their preference for NaPBA or GPB. Compliance with study drug was
`assessed by daily recording of missed doses by the subject and
`monitoring drug accountability records and inspection of the returned
`bottles and vials. Safety was assessed through standard safety
`laboratory tests, physical exams, serial triplicate ECG, and collection
`
`of adverse events. Efficacy was assessed by serial measurement of
`venous ammonia. An independent Data and Safety Monitoring Board
`(DSMB) was chartered to oversee the conduct of the study and an
`interim analysis of safety, ammonia, and PK data was conducted after
`6 subjects completed the study.
`
`2.2. Biochemical analyses
`
`NaPBA and GPB metabolites including PBA, PAA, and PAGN were
`measured by validated liquid chromatography tandem mass spec-
`trometry methods at the bioanalytical
`laboratory, Quest Pharma
`Services. Venous ammonia was measured by the accredited hospital
`laboratory at each site; plasma amino acids were measured by Baylor
`Medical Genetics Laboratories.
`
`2.3. Pharmacokinetic and ammonia sampling
`
`Blood samples for analysis of venous ammonia, intact GPB, and
`NaPBA and GPB metabolites were collected on the last day of dosing
`with either NaPBA or GPB at time zero (pre dose and pre-breakfast)
`and 4, 8, 12, 16, 20 and 24 h post-first dose. Lunch and dinner typically
`were eaten after the 4 and 8 h collections, respectively. Plasma amino
`acids were collected at time 0 (fasting) on days 7 and 14. Urine was
`collected in aliquots of 0–12 h (beginning with the first dose of the
`day) and 12–24 h.
`
`2.4. Pharmacokinetic analyses
`
`Pharmacokinetic parameters of PBA, PAA and PAGN in plasma and
`urine were calculated using a validated version of WinNonlin®
`Enterprise (Version 5.2). Statistical analyses were performed using
`WinNonlin v.5.2 (LinMix Module). Plasma PK parameters, including
`mean and coefficient of variation (standard deviation [SD], expressed
`as a percentage of the mean), were calculated using actual time-
`concentration profiles for each subject and included the following:
`Area under the concentration vs. time curve from time 0 (pre-dose) to
`24 h (AUC0–24), calculated using the linear trapezoid rule, maximum
`plasma concentration at steady state (Cmaxss), minimum plasma
`concentration at steady state (Cminss), time of maximum plasma
`concentration at steady state (Tmaxss), and apparent clearance at
`steady state (CLss/F) (calculated as Dose/AUC0–24 over the bioavailable
`fraction [F]). The amount of PAGN excreted in urine over 24 h was
`calculated by multiplying urine volume with urinary concentrations.
`Summary tables and figures were generated using WinNonlin®
`AutoPilot™ (Version 1.1.1), a configurable software application that
`works with WinNonlin®, and third-party reporting tools, including
`SigmaPlot® versions 9.01 and 10.1 and Microsoft® Office Word and
`Excel 2003 and 2007.
`
`2.5. Ammonia analyses
`
`The primary efficacy endpoint for the switch over phase of the
`study was 24-hour ammonia AUC (NH324-hour AUC), calculated based
`on the sequence of ammonia concentrations outlined above. An
`imputation algorithm was prespecified in the statistical analysis plan
`to allow for calculation of NH324-hour AUC for subjects with missing
`ammonia values.
`
`2.6. Efficacy endpoints and statistical analyses
`
`The primary efficacy endpoint was predefined as comparison of
`NH324-hour AUC on the last day of NaPBA treatment with the last day of
`GPB treatment. Secondary efficacy endpoints included the maximum
`ammonia concentrations and percentage of abnormal ammonia
`values on the last day of treatment with NaPBA vs. GPB. All subjects
`who received any amount of both study medications were included in
`
`
`Page 2 of 7
`
`Owner Ex. 2018
`Par Pharm. v. Horizon
`IPR2015-01117,-01127
`
`

`
`U. Lichter-Konecki et al. / Molecular Genetics and Metabolism 103 (2011) 323–329
`
`325
`
`Table 1
`Patient demographics.
`
`Patients completing the study
`(N=11)
`
`the intention-to-treat (ITT) population, which was the primary
`population for analysis of efficacy and pharmacokinetic parameters.
`A per-protocol population was also prospectively defined as all
`subjects from the ITT population who (a) exhibited ≥80% compliance
`with study medication during inpatient stays (days 7 and 14), (b) did
`not use sodium benzoate within 48 h prior to days 7 and 14,
`(c) differed with respect to protein intake on study days 7 and 14
`by ≤50%, and (d) had calculable ammonia AUC (as defined above) for
`both days 7 and 14.
`Non-inferiority analysis of GPB to NaPBA with respect to ammonia
`control was prospectively defined. An analysis of variance (ANOVA)
`model for the natural log-transformed NH324-hour AUC was constructed
`with factors for treatment as a fixed effect and subject as a random
`effect. The 90% CI for the difference between GPB and NaPBA means
`(GPB minus NaPBA) on the natural log scale was constructed using the
`least square means from the ANOVA model. The difference and lower
`and upper confidence interval of NH324-hour AUC values were
`exponentiated to express the results as geometric means, ratio of
`geometric means, and corresponding CI on the original scale. Non-
`inferiority was to be concluded if the upper bound of the 90% CI was
`less than or equal to 1.25. Correlates of drug dose were determined for
`both NaPBA and GPB using Spearman rank-order correlations. A
`superiority analysis was predefined for the per-protocol population.
`
`3. Results
`
`3.1. Patient demographics, disposition and compliance
`
`Eleven subjects (age range 6–17; 1 male and 10 females) enrolled
`and all 11 subjects completed the protocol defined study procedures.
`As defined by the DSMB Charter, enrollment was temporarily paused
`after six subjects completed the switchover portion of the study
`pending DSMB approval to complete enrollment. Subject demo-
`graphics are summarized in Table 1. One subject each had arginino-
`succinate synthetase (ASS) and arginosuccinate lyase (ASL)
`deficiency; the remaining 9 subjects had ornithine transcarbamylase
`(OTC) deficiencies. Six subjects had neonatal or infantile onset and the
`remainder had later onset UCD. NaPBA had been prescribed for an
`average (SD) of 74.7 (48.2) months at an average (SD) NaPBA dose of
`12.4 (4.4) g/d (equivalent to an average of 322 mg/kg/d or 10.2 g/m2)
`and received the PBA equivalent dose of GPB (average (SD)=10.8 mL,
`equivalent to an average dose of 0.284 mL/kg/d or 313 mg/kg/d). Five
`subjects received NaPBA tablets and 6 received NaPBA powder during
`the study, and 2 subjects who received NaPBA via an NG tube took
`GPB orally during the study. Compliance with treatment was
`excellent; N98% of all scheduled doses of either NaPBA or GPB were
`apparently taken based on monitoring of vials and bottles.
`Dietary protein prescription at baseline among the 11 subjects
`(mean, SD) was 0.75±0.29 g/kg/d (27.7±9.48 g/day). Compliance
`on study with diet was less uniform than compliance with respect to
`study drug. While two subjects showed N50% variance in protein
`intake on days 7 vs. 14 and were excluded from the per protocol
`analysis, overall total mean protein intake was very similar on NaPBA
`and GPB (0.64±0.35 g/kg/d or 24.35 g/d and 0.61±0.16 g/kg/d or
`23.98 g/d on days 7 and 14 respectively; Table 4), as was the
`distribution of dietary protein throughout the day with both GPB and
`NaPBA at breakfast (4.96±2.497 and 5.46±2.943 g, respectively),
`lunch (6.41±4.759 and 5.98±5.332 g, respectively), and dinner
`(6.74±4.653 and 5.71±5.197 g, respectively).
`
`3.2. Safety and tolerability
`
`During the switch over period of the study all reported AEs were
`categorized as mild except one episode of vomiting graded as
`moderate on GPB, which resolved and was attributed to intercurrent
`illness and deemed unrelated to study drug (Table 2). There were no
`
`1 (9.1%)
`10 (90.9%)
`
`10.2 (3.95)
`
`133.66 (16.900)
`
`41.79 (20.135)
`
`Gender [n (%)]
`Male
`Female
`Age (years) at Screening
`Mean (SD)
`Height (cm)
`Mean (SD)
`Weight (kg)
`Mean (SD)
`UCD Diagnosis [n (%)]
`OTC deficiencya
`ASS deficiencyb
`ASL deficiencyc
`UCD onset [n (%)]
`Neonatal (0 – b=30 days)
`Infantile (N30 days – b=2 years)
`Childhood or adult onset (N2 years)
`Duration of NaPBA treatment (months)
`Mean (SD)
`Median
`Min, Max
`Type of NaPBA [n (%)]
`Powderd
`Tablets
`NaPBA daily dose (mg/kg/day)
`12.41 (4.392)
`Mean (SD)
`10.50
`Median
`8.0–20.0
`Min, Max
`2 (18.2%)
`Subjects with a G-tube
`Average prescribed protein intake during study (g/kg/day)
`Mean (SD)
`0.75 ±0.29
`Patients treated with L-Citrulline (%)
`9 (81.8%)
`
`9 (81.8%)
`1 (9.1%)
`1 (9.1%)
`
`3 (27.3%)
`3 (27.3%)
`5 (45.5%)
`
`74.68 (48.220)
`76.00
`0.5 - 162.0
`
`7 (63.6%)
`4 (36.4%)
`
`a Ornithine transcarbamylase deficiency.
`b Arginosuccinate synthetase deficiency.
`c Arginosuccinate lyase deficiency.
`d One patient was switched from powder to tablets for the study.
`
`episodes of hyperammonemic crisis, predefined as blood ammonia
`exceeding 100 µmol/mL plus signs or symptoms of hyperammonemia
`on either NaPBA or GPB. Each patient was asked by the investigator or
`representative to evaluate his or her drug preference on day 14 of the
`study; all 11 subjects stated a preference for GPB.
`
`3.3. Pharmacokinetic analyses
`
`All 11 patients were considered evaluable for the PK analyses, as
`were all measurable concentration values. Values below the lower limit
`of quantification were treated as zero. Individual plasma metabolite
`
`Table 2
`Adverse events 1.
`
`Preferred term
`
`NaPBA (N=11)
`
`GPB (N=11)
`
`Number of subjects with at least one AE
`Grade 1
`Grade 2
`
`Lymphadenopathy (Grade 1)
`Abdominal pain upper (Grade 1)
`Vomiting (Grade 2)
`Decreased appetite (Grade 1)
`Ear infection (Grade 1)
`Upper respiratory tract infection (Grade 1)
`Cardiac murmur (Grade 1)
`Dermatitis contact (Grade 1)
`
`2 (18.2%)
`2 (18.2%)
`0 (0.0%)
`
`1 (9.1%)
`0 (0.0%)
`0 (0.0%)
`1 (9.1%)
`0 (0.0%)
`0 (0.0%)
`1 (9.1%)
`0 (0.0%)
`
`4 (36.4%)
`3 (27.3%)
`1 (9.1%)
`
`0 (0.0%)
`2 (18.2%)
`1 (9.1%)
`0 (0.0%)
`1 (9.1%)
`1 (9.1%)
`0 (0.0%)
`1 (9.1%)
`
`1 Table reflects number of adverse events reported during 7 days of dosing with sodium
`phenylbutyrate (NaPBA) and 7 days of dosing with glycerol phenylbutyrate (GPB).
`
`
`Page 3 of 7
`
`Owner Ex. 2018
`Par Pharm. v. Horizon
`IPR2015-01117,-01127
`
`

`
`326
`
`U. Lichter-Konecki et al. / Molecular Genetics and Metabolism 103 (2011) 323–329
`
`Table 3
`Sodium phenylbutyrate (NaPBA) and glycerol phenylbutyrate (GPB) plasma metabolite concentrations (ug/mL) *.
`
`Time
`
`PBA
`
`NaPBA
`
`0.843 ±1.28
`b1–3.48
`
`Trough (24 h)
`Mean ±SD
`Range
`Peak (12 h)
`Mean ±SD
`36.8±38.2
`2.28–109
`Range
`Range of single measurements (0–24 h)
`Range
`b1–109
`
`GPB
`
`6.43±16.0
`b1–54.5
`
`74.8±52.6
`4.14–161
`
`b1–161
`
`PAA
`
`NaPBA
`
`0.817 ±1.03
`b1–2.84
`
`68.8 ±47.4
`3.43–148
`
`b1–148
`
`* PBA = phenylbutyric acid; PAA = phenylacetic acid; and PAGN = phenylacetylglutamine.
`
`GPB
`
`9.13 ±18.6
`b1–62.9
`
`78.6 ±65.2
`12.1–244
`
`b1–244
`
`PAGN
`
`NaPBA
`
`4.71 ±3.08
`b1–7.94
`
`72.2 ±28.5
`30.8–113
`
`b1–116
`
`GPB
`
`27.7 ±39.2
`2.63–137
`
`88.8 ±33.8
`32.0–136
`
`2.63–153
`
`values varied widely for both NaPBA and GPB whether drawn at peak
`(12 h), trough (24 h; selected over time zero because of monitored
`dosing) or throughout the course of the day (0–24 h) (Table 3). Intact
`GPB was not detectable in plasma.
`Plasma PK parameters of PBA, PAA and PAGN and urinary PK
`parameters of PAGN are summarized in Table 4 and the 24-hour
`concentration profiles are depicted in Fig. 1. Mean systemic PBA exposure
`(AUC0–24) following GPB administration was ~2.7-fold higher (pb0.01)
`than that observed with NaPBA. However, when compared with NaPBA
`administration, there was less variability in plasma PBA levels during
`dosing with GPB, as reflected by the coefficient of variation for AUC, Cmax
`and Cmin (Table 4). Mean (AUC0–24) systemic PAA and PAGN exposure
`during dosing with GPB as compared with NaPBA did not differ
`significantly, albeit also with directionally greater values on GPB as
`compared with NaPBA. Minimum (Cmin) values of PAA were significantly
`greater during GPB as compared with NaPBA dosing.
`
`Table 4
`PK parameters and ammonia at steady state dosing with sodium phenylbutyrate and
`glycerol phenylbutyrate (Study days 7 and 14).
`
`PK/PD parameters
`
`PBA in plasma mean (CV%)
`AUC0–24 (μg·h/mL)
`Cmaxss (μg/mL)
`Cminss (μg/mL)
`PAA in plasma mean (CV%)
`AUC0–24 (μg·h/mL)
`Cmaxss (μg/mL)
`Cminss (μg/mL)
`PAGN in plasma mean (CV%)
`AUC0–24 (μg·h/mL)
`Cmaxss (μg/mL)
`Cminss (μg/mL)
`PAGN in urine mean (CV%)
`Total excreted 0–24 h (g)
`Recovery of PBA as PAGN (%) Fe0–24 (%)
`Ammonia mean (SD)
`AUC (μmol/L)
`Cmaxss (μmol/L)
`Total number (%) of ammonia values
`above ULN⁎
`Diet
`Actual protein intake (g/d) Mean (SD)
`Actual protein intake (g/kg/d) Mean (SD)
`Judged compliant with diet (days 7, 14)
`Difference in ammonia between GPB and NaPBA
`Mean difference of AUC (μmol/L) (SD)
`Ratio of Geometric means
`90% Confidence interval
`95% Confidence interval
`
`Glycerol
`phenylbutyrate
`(n =11)
`
`NaPBA
`(n=11)
`
`631 (44.9)
`95.6 (42.0)
`1.50 (99.8)
`
`236 (105.2)
`37.4 (101.6)
`0.366 (171.3)
`
`964 (63.6)
`90.5 (69.1)
`2.99 (122.1)
`
`773 (73.3)
`75.1 (64.4)
`0.674 (130.5)
`
`1378 (40.2)
`105 (33.5)
`13.1 (64.9)
`
`1015 (44.7)
`74.8 (37.3)
`4.63 (66.4)
`
`12.5 (56.9)
`66.4 (23.9)
`
`12.5 (51.3)
`69.0 (23.9)
`
`603.8 (187.92)
`47.77 (12.800)
`24 (31.6)
`
`814.6 (322.36)
`55.66 (21.607)
`14 (18.4)
`
`23.98 (9.891)
`0.61 (0.16)
`8/11 (72.7%)
`
`24.35 (12.445)
`0.64 (0.35)
`9/11 (81.8%)
`
`-210.8 (310.89)
`0.781
`(0.609, 1.002)
`(0.575, 1.061)
`
`AUC0–24: Area under the concentration from time 0 (pre-dose) to 24 h, Cmaxss: Maximum
`plasma concentration at steady state, Cminss: Minimum plasma concentration at steady
`state, CV% — coefficient of variation.
`⁎ % abnormal ammonia values presented as mean (SD); the denominator is the total
`number of ammonia values at all time points (76).
`
`Similar to plasma, all urines were judged analyzable. One subject
`had no urine sample for the first interval and another did not void
`during the 12–24 h post-dose collecting interval, therefore their total
`PAGN amount excreted over 24 h (U-PAGN24-hour Excr) was adjusted as
`prospectively detailed in pharmacokinetic analysis plan. 24-hour
`PAGN excretion following GPB treatment was very similar to that
`observed for NaPBA (66% vs. 69% recovery of PBA as urinary PAGN),
`although peak urinary PAGN occurred later in the day during GPB
`treatment as compared with NaPBA treatment (percentage of total
`output from 0 to 12 and 12 to 24 h approximately 45% and 55% for GPB
`vs. 57% and 43% for NaPBA).
`
`3.4. Plasma amino acids
`
`Three patients had glutamine values above the normal range
`(266–746 umol/L) on GPB as compared with 6 patients on NaPBA.
`Mean [SD] plasma glutamine levels were non-statistically significant-
`ly lower on GPB (650.3 [187.3] umol/L) vs. NaPBA (725.1 [204.2]
`umol/L). Mean [SD] plasma branched chain amino acid levels were
`similar with NaPBA vs. GPB (isoleucine=34.1 [20.2] vs. 38.6 [9.2],
`leucine=64.0 [38.3] vs. 68.0 [16.9], and valine=134.5 [65.36] vs.
`112.0 [36.36] on NaPBA and GPB, respectively).
`
`3.5. Blood ammonia
`
`Ammonia levels varied widely, increased several fold during the day,
`peaking at around 8–12 h, and in four samples (3 during NaPBA and 1
`during GPB treatment, respectively) exceeded 100 umol/L in the absence
`of clinical symptoms. Only 2 of 154 blood ammonia values were missing;
`all NH324-hour AUC values were calculable and no imputation was
`required. Average ammonia values tended to be lower on GPB than on
`NaPBA assessed as NH324-hour AUC, average blood ammonia, average
`Cmaxss or the percentage of values above the upper limit of normal (as
`per normal values at the respective study site; range 29–54 μmol/L)
`(Table 4, Figs. 2 and 3), although these differences did not achieve
`statistical significance. GPB was determined to be non-inferior to NaPBA
`with respect to ammonia control based on the pre-specified analysis, as
`the upper boundaries of both the 90% (0.609, 1.002) and 95% (0.575,
`1.061) confidence intervals fell below 1.25. Analysis of ammonia control
`in the pre-specified per-protocol population demonstrated significantly
`lower ammonia (623.1 vs. 897.2), assessed as NH324-hour AUC on GPB as
`vs. NaPBA (pb0.05). This per-protocol analysis on 9 of 11 subjects
`excluded the two with a variance of over 50% in dietary protein intake on
`days 7 and 14.
`
`3.6. Effect of age on PK and ammonia during GPB dosing
`
`PK parameters during steady state dosing with GPB analyzed
`separately for subjects ages 6–11 (n=7) vs. 12–17 (n=4) were similar,
`as were mean (SD) ammonia values assessed as NH324-hour AUC (Table 5).
`
`
`Page 4 of 7
`
`Owner Ex. 2018
`Par Pharm. v. Horizon
`IPR2015-01117,-01127
`
`

`
`U. Lichter-Konecki et al. / Molecular Genetics and Metabolism 103 (2011) 323–329
`
`327
`
`GPB Day 14
`NaPBA Day 7
`LOQ = 1 µg/mL
`
`Fig. 2. Twenty Four Hour Ammonia Values on Sodium Phenylbutyrate and Glycerol
`Phenylbutyrate. Venous ammonia was measured for 24 h following one week of dosing
`with either sodium phenylbutyrate (NaPBA; continuous line) or glycerol phenylbutyrate
`(GPB; dotted line) in 11 pediatric subjects. The top panel depicts mean (SE) ammonia
`concentrations over 24 h. The bottom panel depicts overall ammonia values, where the
`bottom and top of the ‘box’ represent the 25th and 75th percentile of all values, the
`horizontal line within the box represents the mean, the open diamond within the box
`represents the median, and the top and bottom of the lines correspond to the maximum
`and minimum observed values, respectively. The open circles above the box for ammonia
`(24-hours AUC) on GPB represent outliers above the 75th percentile.
`
`4. Discussion
`
`No clinically important safety issues were identified during GPB
`dosing and tolerability was satisfactory. No hyperammonemic crises
`occurred during either NaPBA or GPB treatment. Plasma PBA exposure
`was ~2.7 times greater (p b0.01) during dosing with GPB as compared
`with NaPBA, whereas plasma PAA and PAGN exposure differed by
`
`100
`
`10
`
`1
`
`0.1
`
`100
`
`10
`
`1
`
`0.1
`
`100
`
`(µg/mL)
`
` (µg/mL)
`
`10
`
`(µg/mL)
`
`Mean (SD) Plasma PBA Concentration
`
`Median (SD) Plasma PAA Concentration
`
`Median (SD) Plasma PAGN Concentration
`
`0
`
`2
`
`4
`
`6
`
`8
`
`10
`12
`14
`Nominal Time (h)
`
`16
`
`18
`
`20
`
`22
`
`24
`
`GPB Day 14
`NaPBA Day 7
`LOQ = 1 µg/mL
`
`0
`
`2
`
`4
`
`6
`
`8
`
`10
`12
`14
`Nominal Time (h)
`
`16
`
`18
`
`20
`
`22
`
`24
`
`GPB Day 14
`NaPBA Day 7
`LOQ = 1 µg/mL
`
`1
`
`0
`
`2
`
`4
`
`6
`
`8
`
`10
`12
`14
`Nominal Time (h)
`
`16
`
`18
`
`20
`
`22
`
`24
`
`Fig. 1. Plasma phenylbutyric acid (PBA; top panel), phenylacetic acid (PAA; middle
`panel) and phenylacetylglutamine (PAGN; bottom panel) were measured for 24 h
`following one week of dosing with either sodium phenylbutyrate (NaPBA) or glycerol
`phenylbutyrate (GPB) and are displayed as median ±SD. Times 0 and 24 h correspond
`to just prior to dosing and breakfast.
`
`3.7. Correlates of drug dose and ammonia
`
`Drug dose correlated directly and most consistently and strongly
`with urinary PAGN for both NaPBA and GPB treatments (r=0.866;
`p b0.0001 for both NaPBA and GPB combined) (Table 6). Drug dose
`did not correlate with plasma PBA levels assessed as AUC0–24.
`Drug dose did correlate, albeit less strongly and consistently, with
`plasma PAGN and PAA, assessed as AUC0–24. Ammonia assessed as
`NH324-hour AUC correlated neither with blood levels of PBA, PAA or
`PAGN nor with urinary PAGN output. Blood ammonia Cmax correlated
`positively with glutamine, although the correlation was modest
`(r=0.25) and did not achieve significance in this small sample size.
`
`Fig. 3. Venous ammonia in individual subjects following one week of dosing with either
`sodium phenylbutyrate (NaPBA; left) or glycerol phenylbutyrate (GPB; right). The
`values shown represent time-normalized area under the curve and are displayed as
`mean ±SD. Times 0 and 24 h correspond to just prior to dosing and breakfast.
`
`
`Page 5 of 7
`
`Owner Ex. 2018
`Par Pharm. v. Horizon
`IPR2015-01117,-01127
`
`

`
`328
`
`U. Lichter-Konecki et al. / Molecular Genetics and Metabolism 103 (2011) 323–329
`
`Table 5
`PK parameters and ammonia at steady state dosing with glycerol phenylbutyrate
`(Study day 14) by age range.
`
`PK/PD
`parameters
`
`All subjects
`(n =11)
`
`Ages 6–11
`(n =7)
`
`Ages 12–17
`(n =4)
`
`(Day 14; 23.98±9.891 g/d or 0.61±0.16 g/kg/d) and NaPBA (Day 7;
`24.35±12.445 g/d or 0.64±0.35 g/kg/d).
`One possible explanation for the directionally lower ammonia
`values is the generally higher plasma metabolite levels observed
`during GPB dosing. As summarized in Table 4, GPB treatment resulted
`in higher plasma PBA levels overall, including higher peak and trough
`levels, and similar differences in favor of higher plasma levels for PAA
`and PAGN. Plasma PAA levels were better sustained and directionally
`higher over night, as manifested by higher trough levels and this could
`be causally related to the directionally lower ammonia levels during
`GPB dosing.
`The PAA levels are of interest in that neurological toxicity has been
`reported in cancer subjects administered high doses of PAA
`intravenously; this toxicity was associated with PAA blood levels
`ranging from ~499 to 1285 μg/mL [11,12]. However, PAA blood levels
`observed during steady state dosing with both NaPBA and GPB were
`well below the levels reported by Thibault to be associated with
`neurological symptoms (Table 4).
`Another possible explanation for and contributing factor to the
`ammonia findings may be slower gastrointestinal absorption of PBA
`when delivered as GPB. This is supported by prior PK/PD modeling
`(10) and, in the present study, by the proportion of PAGN excreted
`during the 0–12 and 12–24 h time periods (45% and 55% for GPB vs.
`57% and 43% for NaPBA). A slower gastrointestinal absorption of PBA
`is also indicated by lesser variation in PBA concentrations during GPB
`dosing in this study (Table 4).
`While the number of subjects is small, ammonia and PK findings
`appeared to be generally similar during steady state GPB among
`subjects ages 6–11 and 12–17, suggesting that GPB metabolism and
`effect is not dependent on age, per se, within this range (Table 5).
`The present study underscores the difficulty clinicians face in making
`decisions regarding drug dosing based on blood ammonia. Even under
`the controlled conditions of the present trial,
`including excellent
`compliance with diet and study drug in patients whose ammonia
`values were viewed as well controlled by their physicians prior to
`enrollment, ammonia values on average varied more than 10-fold over
`the course of the day. This observation suggests that random values for
`blood ammonia are of limited utility and that ammonia levels should be
`drawn at a constant time in relation to meals and medication for
`monitoring of treatment. Blood levels of metabolites are also problem-
`atic with respect to therapeutic monitoring. As summarized in Tables 3
`and 4 and depicted in Fig. 1, blood levels of PBA, PAA and PAGN all varied
`widely over the course of the day. Levels at individual time points and
`even peak or trough levels varied considerably among these patients
`(Table 3), suggesting that therapeutic metabolite blood levels when
`collected randomly or at peak or trough time points are difficult if not
`impossible to define. Although PAA and/or PAGN exposure assessed as
`24 h area under the curve and measured with 8 samples over 24 h
`did correlate with dose, the correlation was weak and inconsistent and
`24-hour sampling is impractical in routine practice.
`Urinary output of PAGN,
`in contrast to blood levels of drug
`metabolites, shows promise as a biomarker of dose selection and
`compliance monitoring in pediatric UCD patients and is practical to
`perform in routine clinical practice. This makes theoretical sense,
`since PAGN output is stoichiometrically related to waste nitrogen
`scavenging, and PAGN output, measured over 24 h, correlated
`strongly and positively with drug dose (R= 0.909; p b0.001 for GBP
`and R=0.753; p=0.007 for NaPBA).
`Many metabolic specialists use glutamine levels to monitor treatment
`and adjust drug doses. Among the subjects in this study, who were
`viewed as well-controlled under the supervision of a metabolic specialist
`at entry, glutamine levels decreased, although not statistically signifi-
`cantly, after switching from NaPBA to GBP. Glutamine levels also
`correlated modestly and positively with ammonia. However, the
`correlation did not achieve statistical significance and needs evaluation
`in a larg

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