throbber
NIH Public Access
`Author Manuscript
`Mol Genet Metab. Author manuscript; available in PMC 2014 December 01.
`Published in final edited form as:
`Mol Genet Metab. 2013 December ; 110(4): 446–453. doi:10.1016/j.ymgme.2013.09.017.
`
`ELEVATED PHENYLACETIC ACID LEVELS DO NOT
`CORRELATE WITH ADVERSE EVENTS IN PATIENTS WITH
`UREA CYCLE DISORDERS OR HEPATIC ENCEPHALOPATHY
`AND CAN BE PREDICTED BASED ON THE PLASMA PAA TO
`PAGN RATIO
`
`M. Mokhtarania, G.A. Diazb, W. Rheadc, S.A. Berryd, U. Lichter-Koneckie, A. Feigenbaumf,
`A. Schulzef, N. Longog, J. Bartleyh, W. Berquisti, R. Gallagherj, W. Smithk, S.E.
`McCandlessl, C. Hardingm, D.C. Rockeyn, J.M. Vierlingo, P. Mantryp, M. Ghabrilq, R.S.
`Brown Jrr, K. Dickinsona, T. Moorsa, C. Norrisa, D. Coakleya, D.A. Milikiens, SC Nagamanit,
`C. LeMonsu, B. Leet, and B.F. Scharschmidta
`a Hyperion Therapeutics, 601 Gateway Blvd., STE 200, South San Francisco, CA 94080, USA
`
`b Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences,
`Department of Pediatrics, 1428 Madison Ave., New York, NY 10029, USA
`
`c The Medical College of Wisconsin, 9000 W. Wisconsin Ave., Milwaukee, WI 53226, USA
`
`d University of Minnesota, Minneapolis, 420 Delaware St., SE MMC 75, Minneapolis, MN 55455,
`USA
`
`e Children's National Medical Center, 111 Michigan Ave., NW #1950, Washington, DC 20010,
`USA
`
`f The Hospital for Sick Children and University of Toronto, Division of Clinical and Metabolic
`Genetics,, 555 University Avenue, Toronto, ON, Canada M5G1X8
`
`g The University of Utah, Division of Medical Genetics, 2C412 SOM, 50 North Mario Capecchi
`Drive, Salt Lake City, UT 94132, USA
`
`h Long Beach Memorial Hospital, 2865 Atlantic Avenue, #104, Long Beach, CA 90806, USA
`
`© 2013 Elsevier Inc. All rights reserved.
`Corresponding author: Masoud Mokhtarani, M.D. VP, Clinical Development & Medical Affairs Hyperion Therapeutics, Inc. 601
`Gateway Blvd., Ste. 200 South San Francisco, CA 94080 Phone: 1-650-745-7838 Mobile: 1-650-438-8667 Fax: 1-650-745-3581
`Masoud.mokhtarani@hyperiontx.com.
`Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our
`customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of
`the resulting proof before it is published in its final citable form. Please note that during the production process errors may be
`discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
`Conflict of Interest Statement: D. Coakley, K. Dickinson, M. Mokhtarani, T. Moors, C. Norris and B.F. Scharschmidt are/were
`employees of Hyperion at the time of the study. D. Milikien is an employee of Accudata, which was paid by Hyperion to perform the
`biostatistical analyses.
`None of the other authors have a financial interest in Hyperion, although payments were made by Hyperion to all investigators for
`services related to the clinical trials.
`ClinicalTrials.gov Identifiers: ClinicalTrials.gov NCT00551200, NCT00947544, NCT00992459, NCT00947297, NCT00999167,
`NCT 01347073
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Par Pharmaceutical, Inc. Ex. 1039
`Par v. Hyperion, IPR2015-01117
`Page 1 of 21
`
`

`
`Mokhtarani et al.
`
`Page 2
`
`i Stanford University, 750 Welch Road, #116, Palo Alto, CA 94305, USA
`
`j Children's Hospital Colorado, 13123 East 16th Avenue, B153, Aurora, CO 80045, USA
`
`k Maine Medical Center, 1577 Congress Street, 2nd Floor, Portland, ME 04102, USA
`
`l Center for Human Genetics, Case Western Reserve University and University Hospitals Case
`Medical Center, 11100 Euclid Avenue Cleveland, OH 44106, USA
`
`m Oregon Health Sciences University, 3181 SW Sam Jackson Park Road, A036/B198, Mailcode
`L103, Portland, OR 97239, USA
`
`n Medical University of South Carolina, Department of Medicine, 96 Jonathan Lucas St. 803CSB
`Charleston, SC 29425, USA
`
`o Baylor College of Medicine, Departments of Medicine and Surgery, 6620 Main St # 1475,
`Houston, TX 77030, USA
`
`p Liver Institute at Methodist Dallas Medical Center, Dallas 1411 N. Beckley Ave., Pavilion III,
`STE 268 Dallas, TX 75203, USA
`
`q Indiana University, Gastroenterology and Hepatology, Regenstrief Health Center, Room 4100
`1050 Wishard Blvd. Indianapolis, IN 46202, USA
`
`r Columbia University Medical Center, Department of Surgery, Room PH 14, 105, 622 West 168th
`St. New York, NY 10032, USA
`
`s Accudata Solutions, Inc., 886 Oak St., Lafayette, CA 94549, USA
`
`t Baylor College of Medicine, One Baylor Plaza, Room R814, Houston, TX, USA
`
`u The National Urea Cycle Disorders Foundation, Pasadena, CA, 75 South Grand Avenue,
`Pasadena, CA, 91105
`
`Abstract
`
`Background—Phenylacetic acid (PAA) is the active moiety in sodium phenylbutyrate (NaPBA)
`and glycerol phenylbutyrate (GPB, HPN-100), both are approved for treatment of urea cycle
`disorders (UCDs) - rare genetic disorders characterized by hyperammonemia. PAA is conjugated
`with glutamine in the liver to form phenylacetyleglutamine (PAGN), which is excreted in urine.
`PAA plasma levels ≥500 μg/dL have been reported to be associated with reversible neurological
`adverse events (AEs) in cancer patients receiving PAA intravenously. Therefore, we have
`investigated the relationship between PAA levels and neurological AEs in patients treated with
`these PAA pro-drugs as well as approaches to identifying patients most likely to experience high
`PAA levels.
`
`Methods—The relationship between nervous system AEs, PAA levels and the ratio of plasma
`PAA to PAGN were examined in 4683 blood samples taken serially from: [1] healthy adults [2],
`UCD patients ≥2 months of age, and [3] patients with cirrhosis and hepatic encephalopathy (HE).
`The plasma ratio of PAA to PAGN was analyzed with respect to its utility in identifying patients
`at risk of high PAA values.
`
`Mol Genet Metab. Author manuscript; available in PMC 2014 December 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Par Pharmaceutical, Inc. Ex. 1039
`Par v. Hyperion, IPR2015-01117
`Page 2 of 21
`
`

`
`Mokhtarani et al.
`
`Page 3
`
`Results—Only 0.2% (11) of 4683 samples exceeded 500 ug/ml. There was no relationship
`between neurological AEs and PAA levels in UCD or HE patients, but transient AEs including
`headache and nausea that correlated with PAA levels were observed in healthy adults. Irrespective
`of population, a curvilinear relationship was observed between PAA levels and the plasma
`PAA:PAGN ratio, and a ratio > 2.5 (both in μg/mL) in a random blood draw identified patients at
`risk for PAA levels > 500 μg/ml.
`
`Conclusions—The presence of a relationship between PAA levels and reversible AEs in healthy
`adults but not in UCD or HE patients may reflect intrinsic differences among the populations
`and/or metabolic adaptation with continued dosing. The plasma PAA:PAGN ratio is a functional
`measure of the rate of PAA metabolism and represents a useful dosing biomarker.
`
`Keywords
`BUPHENYL; glycerol phenylbutyrate; HPN-100; neurological adverse events; pharmacokinetics;
`RAVICTI; sodium phenylbutyrate
`
`INTRODUCTION
`
`Glycerol phenylbutyrate, a sodium- and sugar-free phenylbutyrate derivative, and sodium
`phenylbutyrate are approved as ammonia lowering agents in patients with urea cycle
`disorders (UCDs). Both are pro-drugs of phenylacetic acid (PAA), which is formed by beta-
`oxidation from phenylbutyric acid (PBA) delivered either as glycerol phenylbutyrate
`following its intestinal hydrolysis by pancreatic lipases [1] or as sodium phenylbutyrate
`following dissociation in the stomach. PAA is conjugated with glutamine by glutamine-N-
`phenylacetyltransferase, largely in the liver and to a lesser extent in the kidney [2], to form
`phenylacetylglutamine (PAGN), which is excreted in urine, thereby providing an alternate
`pathway to urea for waste nitrogen excretion. In controlled studies population
`pharmacokinetic analyses of sodium phenylbutyrate and glycerol phenylbutyrate, it has been
`shown that the gastrointestinal absorption of PBA is approximately 75% slower when
`delivered as glycerol phenylbutyrate vs. sodium phenylbutyrate and that plasma PAA and
`PAGN levels show less variability during glycerol phenylbutyrate dosing. [3]-[7]. There are
`over 30 reports of the administration of sodium phenylacetate or sodium phenylbutyrate to
`healthy volunteers, patients with UCDs or other metabolic disorders and patients with
`cancer, many of which reported some adverse events (AEs) attributed to PAA
`(Supplemental Table 1) [8]-[36]. These reversible AEs in cancer patients were reported in
`studies involving continuous or intermittent intravenous administration designed to maintain
`high levels of PAA, suggesting that duration of exposure as well as peak PAA levels are
`important [35],[3].
`
`The AEs reportedly associated with high levels of PAA have most commonly included
`nausea, headache, emesis, fatigue, weakness, lethargy, somnolence, dizziness, slurred
`speech, memory loss, confusion, and disorientation [35], [36]. Except for the symptoms of
`Kussmaul respiration, metabolic acidosis, cerebral edema, and coma associated with a fatal
`overdose of sodium phenylacetate/sodium benzoate (AMMONUL®)[13], the symptoms
`were rapidly reversible with reduced dosing or interruption of dosing.
`
`Mol Genet Metab. Author manuscript; available in PMC 2014 December 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Par Pharmaceutical, Inc. Ex. 1039
`Par v. Hyperion, IPR2015-01117
`Page 3 of 21
`
`

`
`Mokhtarani et al.
`
`Page 4
`
`Based on a detailed analysis of the timing of the AEs in relation to blood PAA
`concentrations, Simell calculated the safe upper PAA concentration limit to be 3.5 mmol/L,
`equivalent to 476 μg/mL [22], and Thibault reported that AEs were associated with PAA
`levels ranging from 499–1285 μg/mL [35], [36] .
`
`Sodium phenylbutyrate (BUPHENYL®) has been used for over three decades in the
`treatment of UCDs. Despite the fact that the AEs reportedly associated with elevated plasma
`PAA levels can mimic those associated with hyperammonemia, little is known regarding the
`relationship between PAA levels and AEs in UCD patients. The clinical trials of glycerol
`phenylbutyrate (RAVICTI®, HPN-100), which included over 100 UCD patients, 80 of
`whom underwent comparative study of sodium phenylbutyrate and glycerol phenylbutyrate
`[3] - [6] (the largest prospectively studied group of patients with this rare disorder), 193
`patients with advanced cirrhosis complicated by hepatic encephalopathy (HE) [37], and
`more than 90 healthy adult subjects have afforded a unique dataset and opportunity to
`systematically examine the relationship between PAA levels and AEs and to explore
`biomarkers indicative of patients most likely to experience elevated PAA levels.
`
`METHODS
`
`Clinical Studies (Table 1)
`
`Data from a thorough QTc study in healthy adults, five clinical studies in UCD patients and
`an open label safety and dose escalation study as well as a randomized, double-blinded
`controlled phase 2 study of patients with decompensated cirrhosis complicated by HE
`formed the basis for these analyses.
`
`UCD Patients
`
`Eighty UCD patients completed 4 short-term (10 to 28 days) cross-over studies of sodium
`phenylbutyrate vs. glycerol phenylbutyrate (Table 1). The short-term UCD study population
`included 26 pediatric patients ages ≥2 mos through 17 years who received a mean (range)
`dose of 8 (1-19) g/day of glycerol phenylbutyrate or an equivalent dose of sodium
`phenylbutyrate and 54 adults patients ages 18 years or older who received a mean (range)
`dose of 13 (2-34) g/day of glycerol phenylbutyrate or an equivalent dose of sodium
`phenylbutyrate [3] [4] [5][6]. In addition, data from 100 UCD patients enrolled in 12-month
`glycerol phenylbutyrate treatment protocols including 49 children and 51 adults were
`analyzed in relation to PAA levels over time and the occurrence of the symptoms reported in
`cancer patients by Thibault [35][36][4] [5][6] during 12 months treatment.
`
`Patients with Cirrhosis and HE
`
`Data from a 4-week safety and dose escalation study and a multicenter, randomized placebo-
`controlled study of 178 patients with cirrhosis and hepatic encephalopathy who received
`13.2 g/day of glycerol phenylbutyrate (N=90) or placebo (N=88) for 16 weeks were
`analyzed [37], [38] (Table 1). Patients were monitored for safety and frequent PK samples
`were taken over the course of the study.
`
`Mol Genet Metab. Author manuscript; available in PMC 2014 December 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Par Pharmaceutical, Inc. Ex. 1039
`Par v. Hyperion, IPR2015-01117
`Page 4 of 21
`
`

`
`Mokhtarani et al.
`
`Healthy Adults
`
`Page 5
`
`A total of 98 healthy adults (mean age of 28; 53 male 45 female) participated in a blinded,
`randomized, cross over study to assess effects of glycerol phenylbutyrate and its metabolites
`on QTc and other ECG parameters (Table 1). In this protocol 12 subjects received 29.7 g/
`day, 4 subjects 39.6 g/day of glycerol phenylbutyrate and 68 subjects received placebo,
`moxifloxacin as the positive control and glycerol phenylbutyrate at doses of 13.2 g/day and
`19.8 g/day administered three times daily for 3 days.
`
`Adverse Event Mapping
`
`All treatment emergent adverse events (AEs) coded as to Body System as Nervous System
`Disorders using the Medical Dictionary for Regulatory Activities (MedDRA) in subjects
`enrolled in these studies were included in the analyses. For UCD patients, the specific
`toxicities reported by Thibault [35], [36], including nausea, headache, emesis, fatigue,
`weakness, lethargy, somnolence, dizziness, slurred speech, memory loss, confusion, and
`disorientation, exacerbation of neuropathy, pedal edema, hearing loss, abnormal taste,
`arrhythmia, rash, Kussmaul respiration, metabolic acidosis, increased anion gap, tachypnea,
`abdominal discomfort, cerebral edema, and obtundation or coma, were mapped to the
`MedDRA preferred terms in the clinical trial databases.
`
`Analysis of AEs in Relation to PAA Levels
`
`Analyses were based on (a) 2126 samples from 98 healthy adults, (b) 1281 blood PAA and
`PAGN values derived from 80 UCD patients during the short term-switchover studies who
`received both sodium phenylbutyrate and glycerol phenylbutyrate, and (c) 428 samples from
`90 patients with cirrhosis and HE who received glycerol phenylbutyrate. Because plasma
`PAA levels were not always available at the time the patient was experiencing an AE, the
`following rules were applied to associate an AE to a known PAA level. For healthy subjects,
`maximum PAA values recorded after the first dose but within 24 hours of the last dose and
`the incidence of neurological AEs (yes/no) were summarized by dosing period; for periods
`where subjects received placebo or moxifloxacin, the PAA levels were set to 0. For UCD
`patients, maximum PAA values (Cmax) recorded during each dosing period and the
`incidence of neurological AEs were summarized by treatment (glycerol phenylbutyrate or
`sodium phenylbutyrate). For HE patients, each AE was attributed to the PAA result that was
`closest in time to the AE.
`
`The contribution of a 20 μg/mL increase in PAA levels to the probability of a neurological
`AE regardless of relationship to the study drug was examined using Generalized Estimating
`Equations [39]. For healthy subjects, data were summarized for each dose group. Since
`UCD patients received a range of doses, data were summarized for patients receiving a dose
`greater or less than the median dose (equivalent to 11.7 g/day). For HE patients,
`neurological AEs were examined both in relation to blinded treatment group assignment; i.e.
`glycerol phenylbutyrate or placebo, as well as in relation to PAA levels among patients
`treated with glycerol phenylbutyrate.
`
`Mol Genet Metab. Author manuscript; available in PMC 2014 December 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Par Pharmaceutical, Inc. Ex. 1039
`Par v. Hyperion, IPR2015-01117
`Page 5 of 21
`
`

`
`Mokhtarani et al.
`
`Page 6
`
`Analysis of PAA in Relation to Plasma PAA:PAGN Ratio
`
`GEE were used to model the predictive value of plasma PAA:PAGN ratio in identifying
`patients at risk of a high plasma PAA level as defined to have a PAA level equal or greater
`than 400 μg/mL or 500 μg/mL during 24 hours of dosing . Plasma PAA:PAGN ratios were
`grouped into binary categorical range of less than 2.5 or greater than 2.5. The repeated
`measures categorical outcome was modeled using GEE with a logit link function, ratio
`category as the independent variable, and the individual subject ID as the repeated measures
`factor. Confidence intervals for the predicted probabilities were computed by bootstrap
`estimation of 1000 re-samplings of the original data, as detailed in Davison and Hinkley
`[40].
`
`RESULTS
`
`UCD patients (Table 2, Figure 1)
`
`Common AEs reported by at least 10% of patients during short-term treatment with either
`drug included diarrhea, flatulence, and headache. Neurological AEs reported by more than 1
`UCD patient included headache, dizziness and dysgeusia. The mean (SD) PAA Cmax was
`similar in patients who reported at least one neurological AE, as compared with those who
`did not (50.8 (34.5) μg/mL vs 51.5 (49.23) μg/mL respectively). There was no statistically
`significant relationship in UCD patients between the presence or absence of neurological
`AEs and PAA levels during either glycerol phenylbutyrate or sodium phenylbutyrate
`treatment. The odds ratio of a neurological AE occurring for each 20 μg/mL increase in
`PAA levels for the two drugs combined, controlling for dose level, was 0.929, very close to
`1 indicating that increasing levels of PAA were not associated with an increase in
`neurological AEs in these studies. There was no difference in the frequency of the PAA-
`associated AEs reported in cancer patients by Thibault [35], [36] in adult vs. pediatric UCD
`patients in the short-term controlled studies, despite the generally higher PAA levels in
`pediatric patients (Supplemental Table 2).
`
`A total of 100 UCD patients enrolled in 12-month studies of glycerol phenylbutyrate
`received a mean (SD) total dose of 11.01 (5.970) g/day (range: 0.8–34.3 g). Overall common
`AEs reported in at least 10% of UCD patients during long-term treatment included vomiting,
`upper respiratory tract infection, nausea, nasopharyngitis, diarrhea, headache,
`hyperammonemia, decreased appetite, cough, fatigue, dizziness, and oropharyngeal pain.
`There was no increase either in plasma PAA levels (Supplemental Figure 1) or the rate of
`AEs over time. Just as in the short-term studies there was no difference between pediatric
`and adult patients in the frequency of the PAA-associated AEs reported in cancer patients by
`Thibault (Supplemental Table 2).
`
`Patients with cirrhosis and HE (Table 2, Figure 1)
`
`Of 88 patients randomized to placebo, 48.9% reported a neurological AE as compared to
`40.9% of 90 patients randomized to glycerol phenylbutyrate. Of the 428 PAA data points
`from patients randomized to glycerol phenylbutyrate, 46 were in patients who reported a
`neurological AE and 382 in patients who did not. The mean (SD) PAA value closest to
`occurrence of an AE was 61.4 (75.3) μg/mL while the mean PAA value not temporally
`
`Mol Genet Metab. Author manuscript; available in PMC 2014 December 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Par Pharmaceutical, Inc. Ex. 1039
`Par v. Hyperion, IPR2015-01117
`Page 6 of 21
`
`

`
`Mokhtarani et al.
`
`Page 7
`
`associated with an AE was 36.4 (55.6) μg/mL (p=0.77) (Figure 2). Similar to UCD patients,
`there was no increase in the odds of experiencing a neurological AE with each 20 μg/mL
`increment in PAA levels in cirrhosis patients (odds ratio 1.086; p=0.172) indicating that at
`the dose of 13.2 g/day the odds of experiencing a neurological AE did not increase with an
`increase in PAA level.
`
`Healthy subjects
`
`Common AEs in ≥ 10% of healthy volunteers included headache, nausea, and dizziness.
`Neurological AEs increased in frequency with increasing dose, ranging from 26.5% for 13.2
`g/day to 91.7% for 29.7 g/day. Among those who reported a neurological AE, PAA values
`were higher for the 19.8 g/day, 29.7 g/day, and 39.6 g/day dosing periods than for the 13.2
`g/day dosing period (Figure 2, Table 2). PAA levels increased as the dose of glycerol
`phenylbutyrate increased. In the case of the 13.2 g/day dose group, the difference was
`statistically significant (73.3 vs. 41.6, p <0.001) (Table 2). Logistic regression analysis
`indicated that each increment in PAA of 20 μg/mL was associated with increasing odds of
`experiencing a neurological AE (odds ratio = 1.75; p = 0.006). Individual AEs reported by
`healthy adults were generally transient and typically began within 36 hours of dosing and
`generally resolved with continued dosing, as depicted in Supplemental Figure 2.
`
`Plasma PAA:PAGN ratio as a Predictor of Elevated PAA Levels
`
`PAA levels showed considerable variation over a 24-hr period in all patients regardless of
`the dose, drug and population (Figure 3). Unlike PAA, the ratio of PAA:PAGN was
`comparatively constant over 24 hours (data not shown). A curvilinear relationship was
`observed between PAA and PAA:PAGN in all populations, with a sharp upward inflexion
`beginning with PAA concentrations approaching 200 μg/ml and a PAA:PAGN of
`approximately 2.5 or greater (Figure 4). Only 11 of a total of 4683 samples exceeded the
`500 ug/ml threshold level reported by Thibault to be associated with occurrence of
`neurological AEs in cancer patients. The estimated probabilities of correctly detecting a ratio
`≥2.0 based on a single plasma sample taken at any time between the fasting morning sample
`(0 hr time point) and early evening (12 hr time point) remained relatively constant (77% to
`84%), indicating that the timing of blood draw did not have an impact on the ratio of
`PAA:PAGN in plasma regardless of the PAA concentration. Patients with a ratio ≥2.5 had
`significantly higher PAA levels than those with a ratio ≤2.5 (p<0.0001) and PAA:PAGN
`ratios ≥2.5 had an approximately 20 times higher probability of being associated with PAA
`levels > 400 μg/ml (0.8% vs. 19.1%) or 500 μg/ml (0.3% vs. 8.4%) (Table 3).
`
`DISCUSSION
`
`No relationship was observed among UCD patients between PAA levels and either
`neurological AEs, or the specific AEs reported by Thibault, during treatment with either
`glycerol phenylbutyrate or sodium phenylbutyrate. This is supported by (a) the absence of a
`relationship during short term treatment in UCD patients, in which the odds ratio for the
`likelihood of a neurological AE for every 20 μg/mL increase in PAA levels was 0.929, (b)
`the absence of a difference in the frequency of AEs similar to those reported in cancer
`patients by Thibault between pediatric and adult UCD patients during short or long-term
`
`Mol Genet Metab. Author manuscript; available in PMC 2014 December 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Par Pharmaceutical, Inc. Ex. 1039
`Par v. Hyperion, IPR2015-01117
`Page 7 of 21
`
`

`
`Mokhtarani et al.
`
`Page 8
`
`treatment, despite generally higher PAA levels in pediatric patients, and (c) the absence of
`any change in either PAA levels or the pattern of AEs during 12 months of dosing.
`Similarly, no statistical relationship was noted between PAA levels and neurological AEs
`among HE patients treated with 13.2g/day of glycerol phenylbutyrate for 16 weeks, as there
`was no difference in neurological AEs between the glycerol phenylbutyrate and placebo
`treatment arms, nor was there a relationship between PAA levels and the occurrence of
`neurological AEs.
`
`Among the healthy adult volunteers, a relationship was observed between PAA levels and
`the occurrence of any neurological AE (e.g. headache, dizziness, vomiting and nausea).
`These AEs were generally mild, started early in the dosing period, and disappeared with
`continued dosing. The theoretical risk of PAA toxicity is expected to be similar for sodium
`phenylbutyrate or glycerol phenylbutyrate, as both drugs convert to PAA upon absorption.
`The AEs reported by healthy volunteers in these studies receiving glycerol phenylbutyrate
`are generally consistent with prior reports involving administration of sodium
`phenylbutyrate. The mechanism for these AEs is unknown, although interference with brain
`biochemical function has been suggested [41]. These differences between populations may
`be attributable either to metabolic differences between UCD and HE patients, who exhibit
`pathological nitrogen retention and high glutamine levels, as compared with healthy adults,
`and/or metabolic adaptation that may occur with continued exposure to PAA in chronically
`treated patients. Consistent with adaptation are the findings that AEs tended to disappear
`with continued dosing in healthy adults and that the UCD patients enrolled in these studies
`had been treated with sodium phenylbutyrate for an average of more than 9 years.
`
`While most human tissues are capable of beta-oxidation and, hence, conversion of
`phenylbutyrate to PAA [42], enzymatic conversion of PAA to PAGN occurs primarily in the
`liver [2]. This may explain why conversion of PAA to PAGN appears to be a rate-limiting
`step in the metabolism of PAA prodrugs and why PAA metabolism may be compromised
`when liver function is poor, when availability of the precursor glutamine may be limited as
`in healthy subjects, and/or when the capacity of the enzymatic conversion may be limited as
`in very young children [7]. Regardless of the reason, decreases in the rate of PAGN
`formation are associated with an increased ratio of PAA to PAGN in plasma. It is interesting
`in this regard that the upward inflexion in PAA values assessed as a function of the
`PAA:PAGN ratio occurs at a concentration similar to the estimated Km of this reaction
`based on population PK modeling, which is approximately 190 μM as previously described
`by Monteleone et al [7].
`
`In clinical practice, interpretation of an individual PAA value is compromised by the fact
`that concentrations vary considerably over the course of the day due to the relatively short
`half-lives of PBA and PAA. For example among the clinical trials comprising the present
`analyses, plasma PAA fluctuation index varied from 843% - 3931%; and fasting and
`maximal PAA levels in HE patients ranged from 0 - 1.3 μg/mL and 248 - 532 μg/mL,
`respectively. As compared with measurement of PAA alone, measurement of the
`PAA:PAGN ratio appears to be a useful proxy for the efficiency with which an individual
`patient converts PAA to PAGN, and a predictor of patients at risk of having an elevated
`PAA level. The PAA:PAGN ratio also has an important clinical advantage in that it remains
`
`Mol Genet Metab. Author manuscript; available in PMC 2014 December 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Par Pharmaceutical, Inc. Ex. 1039
`Par v. Hyperion, IPR2015-01117
`Page 8 of 21
`
`

`
`Mokhtarani et al.
`
`Page 9
`
`comparatively constant over the day and, therefore, is more readily interpretable in a random
`blood draw.
`
`These analyses have several limitations. First, although pharmacokinetic and safety data
`were derived from controlled prospective studies, the analyses of the frequency of the
`specific AEs reported by Thibault et al. [35], [36] were done as post hoc analysis. Second,
`PAA levels were not always available at precisely the time of occurrence of neurological
`AEs, though a conservative approach was taken in these analyses by utilizing the highest
`recorded PAA for that dosing period. Finally, these conclusions pertaining to the absence of
`a statistical relationship between plasma PAA levels and neurological AEs apply at the
`population levels and may not apply to individual UCD or HE patients [38]. Since the
`symptoms reportedly associated with elevated PAA levels are non-specific and similar to
`those associated with elevated ammonia, it is possible that PAA may occasionally cause
`reversible AEs that go clinically unrecognized or are attributed to something else.
`
`Collectively, the present findings indicate that the PAA:PAGN ratio is a useful dosing
`biomarker suitable for use with random blood draws and they suggest further that dose
`reduction may be warranted in patients receiving PAA prodrugs with an elevated plasma
`PAA:PAGN ratio who exhibit neurological adverse events not explained by elevated
`ammonia or intercurrent illness.
`
`Supplementary Material
`
`Refer to Web version on PubMed Central for supplementary material.
`
`Acknowledgments
`
`The authors gratefully acknowledge and thank the efforts of the Study Coordinators and nursing staff who made
`these trials possible, including D. Bartholomew (Nationwide Children's Hospital), S. Cederbaum, D. Wong
`(University of California), J. Vockley (Children's Hospital of Pittsburgh), S. Bart, M. Al-Ibraham (SNBL), M.S.
`Korson (Tufts Medical Center), D. Kronn (Westchester Medical Center), R. Zori (University of Florida), J.L.
`Merritt (Seattle Children's Hospital), N. Schrager (Mount Sinai School of Medicine), A. Donovan, J. Crawford,
`Pediatric TRU Staff, K. Defouw, J. Balliet (The Medical College of Wisconsin), M. Keuth, N. O’Donnell (Long
`Beach Memorial Hospital), M. Hussain, E. Bailey, M. Ambreen (The Hospital for Sick Children, University of
`Toronto, ON, Canada), C. Bailey, A. Lang (The University of Utah), J. Perry, V. de Leon, A. Niemi, K. Cusmano
`(Stanford University),T. Carlson, J. Parker, S. Elsbecker (University of Minnesota), K. Simpson (Children's
`National Medical Center), K. Regis (Nationwide Children's Hospital), A. Behrend, T. Marrone, J. Martin (Oregon
`Health Sciences University), N. Dorrani (University of California, Los Angeles), M.B. Frohnapfel, S. Bergant, J.
`Haky, C. Tasi, C. Heggie (Case Western Reserve University), S. Mortenson (Maine Medical Center), S. Deward
`(Children's Hospital of Pittsburgh), S. Burr (Children's Hospital Colorado ), K. Bart, C. Duggan (SNBL), K.
`Murray, C. Dedomenico (Tufts Medical Center), C. Gross (University of Florida), L. Brody (Seattle Children's
`Hospital), M. Mullins, S. Carter, A. Tran, J. Stuff, TCH General Clinical Research Center nursing staff (Baylor), B.
`McGuire (University of Alabama), D. Wolf (New York Medical College), C. O’Brien (University of Miami), R.
`O'shea (Cleveland Clinic), I. Zupanets (National University of Pharmacy of MH of Ukraine), Kathy Lisam
`(Hyperion), as well as the Clinical and Translational Science Awards/General Clinical Research Center Grants
`(Baylor College of Medicine, M01RR00188; Case Western Reserve University, UL1RR024989; Clinical and
`Translational Science Institute at Children's National Medical Center NIH/NCRR, UL1RR31988; Medical College
`of Wisconsin, UL1RR31973; Mount Sinai School of Medicine, UL1RR29887; Oregon Health & Science
`University, UL1RR24140; Stanford University, UL1RR25744; Tufts University, UL1RR25752; University of
`California, Los Angeles, UL1RR33176; University of Colorado, UL1RR25780; University of Florida,
`UL1RR29890; University of Minnesota, UL1RR33183; University of Pittsburgh, UL1RR24153, UL1TR000005;
`University of Utah, UL1RR25764; University of Washington, UL1RR25014), the Urea Cycle Disorders
`Consortium (NIH Grant U54RR019453) and grants from the O’Malley Foundation and Kettering Fund which
`provided support. SCS. Nagamani is an awardee of the National Urea Cycle Disorders Foundation Research
`Fellowship. The authors also thank G. Enns (Stanford) for his comments on the manuscript.
`
`Mol Genet Metab. Author manuscript; available in PMC 2014 December 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Par Pharmaceutical, Inc. Ex. 1039
`Par v. Hyperion, IPR2015-01117
`Page 9 of 21
`
`

`
`Mokhtarani et al.
`
`List of Abbreviations
`
`Page 10
`
`GEE
`
`GPB
`
`HE
`
`NaPBA
`
`PAA
`
`generalized estimating equations
`
`glycerol phenylbutyrate (generic name for glyceryl tri (4-
`phenylbutyrate), also referred to as HPN-100 or RAVICTI®)
`
`hepatic encephalopathy
`
`sodium phenylbutyrate (BUPHENYL®)
`
`phenylacetic acid
`
`PAA:PAGN ratio
`
`ratio of the concentrations in μg/mL of PAA to PAGN in plasma
`
`PAGN
`
`PBA
`
`SE
`
`SO
`
`UCD
`
`REFERENCES
`
`phenylacetylglutamine
`
`phenylbutyric acid
`
`safety extension
`
`switchover
`
`urea cycle disorder
`
`1. McGuire BM, Zupanets I, Lowe ME, Syplyviy V, Monteleone JP, Gargosky S, et al. Pharmacology
`and Safety of a Novel Ammonia Lowering Agents In Healthy Adults and Adults with Cirrhosis.
`Hepatology. 2010; 51:2077–85. [PubMed: 20512995]
`2. Moldave K. A Meister, Synthesis of phenylacetylglutamine by human tissue. J. Biol. Chem. 1957;
`229:463–76. [PubMed: 13491597]
`3. Lee B, Rhead W, Diaz GA, Scharschmidt BF, Mian A, Shchelochkov O, et al. Phase 2 comparison
`of a novel ammonia scavengin

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