throbber
Molecular Genetics and Metabolism 110 (2013) 446–453
`
`Contents lists available at ScienceDirect
`
`Molecular Genetics and Metabolism
`
`j o ur n a l h o m e p a g e : w ww . e l s e v i e r . c om / l o c a t e / y mg m e
`
`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. Mokhtarani a,⁎, G.A. Diaz b, W. Rhead c, S.A. Berry d, U. Lichter-Konecki e, A. Feigenbaum f, A. Schulze f,
`N. Longo g, J. Bartley h, W. Berquist i, R. Gallagher j, W. Smith k, S.E. McCandless l, C. Harding m, D.C. Rockey n,
`J.M. Vierling o, P. Mantry p, M. Ghabril q, R.S. Brown Jr. r, K. Dickinson a, T. Moors a, C. Norris a, D. Coakley a,
`D.A. Milikien s, S.C. Nagamani t, C. LeMons u, B. Lee t, B.F. Scharschmidt a
`a Hyperion Therapeutics, 601 Gateway Blvd., Suite 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 M5G1X8, Canada
`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
`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, USA
`
`a r t i c l e
`
`i n f o
`
`a b s t r a c t
`
`Article history:
`Received 26 September 2013
`Accepted 29 September 2013
`Available online 8 October 2013
`
`Keywords:
`BUPHENYL
`Glycerol phenylbutyrate
`HPN-100
`Neurological adverse events
`RAVICTI
`Sodium phenylbutyrate
`
`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 intravenous-
`ly. 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 of ≥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.
`Results: Only 0.2% (11) of 4683 samples exceeded 500μg/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 be-
`tween PAA levels and the plasma PAA:PAGN ratio, and a ratio N 2.5 (both in μg/mL) in a random blood draw iden-
`tified patients at risk for PAA levels N 500 μg/ml.
`
`Abbreviations: GEE, generalized estimating equations; GPB, glycerol phenylbutyrate (generic name for glyceryl tri (4-phenylbutyrate), also referred to as HPN-100 or RAVICTI®); HE,
`hepatic encephalopathy; NaPBA, sodium phenylbutyrate (BUPHENYL®); PAA, phenylacetic acid; PAA:PAGN ratio, ratio of the concentrations in μg/mL of PAA to PAGN in plasma; PAGN,
`phenylacetylglutamine; PBA, phenylbutyric acid; SE, safety extension; SO, switchover; UCD, urea cycle disorder.
`☆ ClinicalTrials.gov identifiers: ClinicalTrials.gov NCT00551200, NCT00947544, NCT00992459, NCT00947297, NCT00999167, and NCT 01347073.
`⁎ Corresponding author at: VP, Clinical Development & Medical Affairs, Hyperion Therapeutics, Inc., 601 Gateway Blvd., Ste. 200, South San Francisco, CA 94080, USA. Fax: +1 650 745 3581.
`E-mail address: Masoud.mokhtarani@hyperiontx.com (M. Mokhtarani).
`
`1096-7192/$ – see front matter © 2013 Elsevier Inc. All rights reserved.
`http://dx.doi.org/10.1016/j.ymgme.2013.09.017
`
`[6.1.2.9] [Mokhtarani et al - PAA and PAA-PAGN ratio on line publication.pdf] [Page 1 of 8]
`
`Horizon Exhibit 2018
`Lupin v. Horizon
`IPR2018-00459
`
`1 of 8
`
`

`

`M. Mokhtarani et al. / Molecular Genetics and Metabolism 110 (2013) 446–453
`
`447
`
`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 repre-
`sents a useful dosing biomarker.
`
`© 2013 Elsevier Inc. All rights reserved.
`
`1. 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 disso-
`ciation 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 gastrointes-
`tinal absorption of PBA is approximately 75% slower when delivered
`as glycerol phenylbutyrate vs. sodium phenylbutyrate and that plas-
`ma 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 (Sup-
`plemental Table 1) [8–36]. These reversible AEs in cancer patients
`were reported in studies involving continuous or intermittent intrave-
`nous administration designed to maintain high levels of PAA, suggesting
`that duration of exposure as well as peak PAA levels are important
`[3,35].
`The AEs reportedly associated with high levels of PAA have
`most commonly included nausea, headache, emesis, fatigue, weak-
`ness, 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. Based on a de-
`tailed analysis of the timing of the AEs in relation to blood PAA concen-
`trations, 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 pa-
`tients. The clinical trials of glycerol phenylbutyrate (RAVICTI®,
`HPN-100), which included over 100 UCD patients, 80 of whom
`underwent a comparative study of sodium phenylbutyrate and
`glycerol phenylbutyrate [3–6] (the largest prospectively studied
`group of patients with this rare disorder), 193 patients with ad-
`vanced 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 bio-
`markers indicative of patients most likely to experience elevated
`PAA levels.
`
`2. Methods
`
`2.1. Clinical studies (Table 1)
`
`Data from a thorough QTc study in healthy adults, five clinical stud-
`ies 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.
`
`2.2. 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 of ages ≥ 2 mos through 17 years who received a mean
`(range) dose of 8 (1–19) g/day of glycerol phenylbutyrate or an equiva-
`lent 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 [3–6]. 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 [4–6,35,36] during
`12 months treatment.
`
`2.3. Patients with cirrhosis and HE
`
`Data from a 4-week safety and dose escalation study and a multicen-
`ter, 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). The patients were monitored for safety and
`frequent PK samples were taken over the course of the study.
`
`2.4. Healthy adults
`
`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 pa-
`rameters (Table 1). In this protocol 12 subjects received 29.7g/day, 4 sub-
`jects 39.6 g/day of glycerol phenylbutyrate and 68 subjects received
`placebo, moxifloxacin as the positive control and glycerol phenylbutyrate
`at doses of 13.2g/day and 19.8g/day administered three times daily for 3
`days.
`
`2.5. 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 the 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 neu-
`ropathy, pedal edema, hearing loss, abnormal taste, arrhythmia,
`rash, Kussmaul respiration, metabolic acidosis, increased anion gap,
`tachypnea, abdominal discomfort, cerebral edema, and obtundation or
`
`[6.1.2.9] [Mokhtarani et al - PAA and PAA-PAGN ratio on line publication.pdf] [Page 2 of 8]
`
`2 of 8
`
`

`

`448
`
`M. Mokhtarani et al. / Molecular Genetics and Metabolism 110 (2013) 446–453
`
`Table 1
`Clinical studies and subject disposition.
`
`Populations
`
`Study ID, design and objectives
`
`Healthy adult volunteers
`
`HPN-100-010
`Thorough QT/QTc study
`Arm 1: safety run-in
`Arm 2: double-blind, randomized, crossover
`
`UCD patients
`
`UP 1204-003
`Phase 2, open-label, fixed-sequence, switch-over study
`
`HPN-100-006
`Phase 3, randomized, double-blind, crossover,
`active-controlled, multiple-dose study
`HPN-100-005SO
`Phase 2, open-label, fixed-sequence, switch-over,
`multiple-dose study with 12-month safety extension
`HPN-100-012SO
`Phase 3b, open-label, fixed-sequence, switch-over study
`
`HPN-100-005SE
`Phase 2, open-label 12-month
`safety-extension study
`HPN-100-012SE
`Phase 2, open-label 12-month safety-extension study
`
`HPN-100-007
`Phase 3, open-label, 12-month safety-extension study
`
`HE Patients
`
`HPN-100-008 (Part A)
`open label, safety and dose-escalation study
`HPN-100-008a
`randomized, double-blind, placebo-controlled phase 2 study
`
`Ages/no. treated
`Adults ≥ 18 N:98
`Arm 1: 9 mL TID: 12
`12 mL TID: 4
`Arm 2: 4 mL TID: 68
`6 mL TID: 68
`Adults ≥ 18
`N:10
`
`Adults ≥ 18
`N: 44
`
`Pediatric, patients ages 6–17
`N:11
`
`Pediatric, patients ages 2 months
`to b6 yrs
`N:15
`Pediatric ages
`6–17 years
`N: 17
`Pediatric ages
`29 days to b6 years
`N: 23
`Adult and pediatric
`ages ≥ 6
`N: 60 (51 adults, 9 pediatric patients)
`Adults ≥ 18
`N: 15
`Adults ≥ 18
`N: 178
`
`Study drug/Duration
`
`GPB
`3 days
`
`Open label, fixed sequence, NaPBA to
`GPB switchover
`7 days on each drug
`GPB and NaPBA
`14 days randomized, double blind,
`double dummy cross over
`Open label, fixed sequence, NaPBA to
`GPB switchover
`7 days on each drug
`Open label, fixed sequence, NaPBA to
`GPB switchover
`≤7 days on each drug
`GPB
`12 months
`
`GPB
`12 months
`
`GPB
`12 months
`
`GPB
`4 weeks
`GPB
`16 weeks
`
`GPB — glycerol phenylbutyrate; HE — hepatic encephalopathy; NaPBA — sodium phenylbutyrate; SE — safety extension; SO — switchover; UCD — urea cycle disorders.
`a Used for analysis of PAA levels in relation to AEs only.
`
`coma, were mapped to the MedDRA preferred terms in the clinical trial
`databases.
`
`2.7. Analysis of PAA in relation to plasma PAA:PAGN ratio
`
`2.6. 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 sub-
`jects, maximum PAA values recorded after the first dose but within 24h
`of the last dose and the incidence of neurological AEs (yes/no) were
`summarized by dosing period; for periods where subjects received pla-
`cebo 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 20μg/mL increase in PAA levels to the probabil-
`ity of a neurological AE regardless of relationship to the study drug
`was examined using generalized estimating equations [39]. For healthy
`subjects, the data were summarized for each dose group. Since the UCD
`patients received a range of doses, the data were summarized for pa-
`tients receiving a dose greater or less than the median dose (equivalent
`to 11.7 g/day). For the HE patients, neurological AEs were examined
`both in relation to the blinded treatment group assignment; i.e. glycerol
`phenylbutyrate or placebo, as well as in relation to PAA levels among
`the patients treated with glycerol phenylbutyrate.
`
`GEE were used to model the predictive value of the 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 h of dosing. The plasma PAA:PAGN ratios were
`grouped into binary categorical ranges of less than 2.5 or greater than
`2.5. The repeated measure 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].
`
`3. Results
`
`3.1. UCD patients (Table 2, Fig. 1)
`
`Common AEs reported by at least 10% of the patients during short-
`term treatment with either drug included diarrhea, flatulence, and
`headache. Neurological AEs reported by more than 1 UCD patient in-
`cluded 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 the 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 in-
`creasing levels of PAA were not associated with an increase in neurolog-
`ical AEs in these studies. There was no difference in the frequency of the
`PAA-associated AEs reported in the cancer patients by Thibault [35,36]
`
`[6.1.2.9] [Mokhtarani et al - PAA and PAA-PAGN ratio on line publication.pdf] [Page 3 of 8]
`
`3 of 8
`
`

`

`M. Mokhtarani et al. / Molecular Genetics and Metabolism 110 (2013) 446–453
`
`449
`
`in adult vs. pediatric UCD patients in the short-term controlled studies,
`despite the generally higher PAA levels in the 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 re-
`spiratory 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).
`
`3.2. Patients with cirrhosis and HE (Table 2, Fig. 1)
`
`Of the 88 patients randomized to placebo, 48.9% reported a neuro-
`logical AE as compared to 40.9% of the 90 patients randomized to glyc-
`erol phenylbutyrate. Of the 428 PAA data points from the patients
`randomized to glycerol phenylbutyrate, 46 were in the patients who re-
`ported a neurological AE and 382 in the 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 associated with an AE was
`36.4 (55.6) μg/mL (p = 0.77) (Fig. 2). Similar to the UCD patients,
`there was no increase in the odds of experiencing a neurological AE
`with each 20 μg/mL increment in PAA levels in the 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.
`
`3.3. Healthy subjects
`
`Common AEs in ≥10% of healthy volunteers included headache,
`nausea, and dizziness. Neurological AEs increased in frequency with in-
`creasing dose, ranging from 26.5% for 13.2g/day to 91.7% for 29.7g/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 (Fig. 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 b 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 h of dosing and generally resolved with
`continued dosing, as depicted in Supplemental Figure 2.
`
`3.4. Plasma PAA:PAGN ratio as a predictor of elevated PAA levels
`
`PAA levels showed considerable variation over a 24-hr period in all
`the patients regardless of the dose, drug and population (Fig. 3). Unlike
`PAA, the ratio of PAA:PAGN was comparatively constant over 24h (data
`not shown). A curvilinear relationship was observed between PAA and
`PAA:PAGN in all the populations, with a sharp upward inflection begin-
`ning with PAA concentrations approaching 200 μg/ml and a PAA:PAGN
`of approximately 2.5 or greater (Fig. 4). Only 11 of a total of 4683 sam-
`ples exceeded the 500 μg/ml threshold level reported by Thibault to be
`associated with the 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 morn-
`ing sample (0 h time point) and early evening (12 h 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 b 0.0001) and PAA:PAGN ratios ≥ 2.5 had an approximately 20
`times higher probability of being associated with PAA levels N 400
`μg/ml (0.8% vs. 19.1%) or 500 μg/ml (0.3% vs. 8.4%) (Table 3).
`
`4. 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 sodi-
`um phenylbutyrate. This is supported by (a) the absence of a relation-
`ship 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 treatment,
`despite generally higher PAA levels in pediatric patients, and (c) the ab-
`sence of any change in either PAA levels or the pattern of AEs during 12
`months of dosing. Similarly, no statistical relationship was noted be-
`tween PAA levels and neurological AEs among HE patients treated
`with 13.2 g/day of glycerol phenylbutyrate for 16 weeks, as there was
`
`Table 2
`Analysis of the relationship between PAA and neurological adverse events.
`
`Healthy subjects
`
`Dose
`
`13.2 g/d
`
`19.8 g/d
`
`29.7 g/d
`
`39.6 g/d
`
`AE Reported
`
`n
`
`Yes
`15
`
`Plasma PAA (μg/mL)
`Mean
`32.9
`SD
`13.1
`Median
`35.5
`p-valuea
`0.076
`1.75b
`Odds ratio
`p-valued
`0.006
`
`No
`51
`
`26.9
`12.2
`24.6
`
`Yes
`31
`
`86.3
`53.9
`73.3
`b0.001
`
`No
`38
`
`48.1
`30.2
`41.6
`
`Yes
`8
`
`211.1
`118.4
`194.8
`0.222
`
`No
`1
`
`65.0
`–
`65.0
`
`Yes
`3
`
`282.4
`179.5
`328.0
`0.500
`
`No
`1
`
`64.8
`–
`64.8
`
`UCD patients
`
`b=11.7 g/d
`
`N11.7 g/d
`
`HE patients
`
`13.2 g/day
`
`No
`75
`
`55.3
`91.4
`24.3
`
`Yes
`11
`
`58.7
`38.2
`70.1
`0.935
`
`No
`68
`
`64.6
`54.1
`48.1
`
`Yes
`4
`
`30.9
`7.4
`30.6
`0.627
`0.929c
`0.529
`
`No
`52
`
`36.4
`55.6
`20.9
`
`Yes
`36
`
`61.4
`75.3
`29.9
`0.77
`1.086c
`0.172
`
`AE — adverse event; HE — hepatic encephalopathy; PAA — phenylacetic acid; UCD — urea cycle disorders.
`UCD patient data are derived from studies UP-1204-003, HPN-100-005SO, HPN-100-006, and HPN-100-012SO. HE patient data are derived from study HPN-100-012 Part B only.
`a p-value comparing subjects reporting a neurological AE within each dose group using an exact non-parametric Mann–Whitney test.
`b Odds ratio of experiencing a neurological AE associated with a 20 μg/mL increase in PAA, Cmax controlling for dose group, including placebo and moxifloxacin, for which PAA was
`assumed to be zero.
`c Odds ratio (p-value) of incidence of neurological AE associated with a 20-μg/mL increase in PAA Cmax.
`d p-value for the odds ratio.
`
`[6.1.2.9] [Mokhtarani et al - PAA and PAA-PAGN ratio on line publication.pdf] [Page 4 of 8]
`
`4 of 8
`
`

`

`450
`
`M. Mokhtarani et al. / Molecular Genetics and Metabolism 110 (2013) 446–453
`
`Experienced neurological AE
`
`No
`
`Yes
`
`Sodium Phenylbutyrate
`
`Experienced neurological AE
`
`No
`
`Yes
`
`Glycerol Phenylbutyrate
`
`Experienced neurological AE
`
`No
`
`Yes
`
`Glycerol Phenylbutyrate
`
`Fig. 1. Lack of relationship between PAA levels and neurological AEs in UCD and HE pa-
`tients. The top and middle panels depict box-and-whisker plots for the mean maximal
`(Cmax) concentration of PAA during dosing of UCD patients with sodium phenylbutyrate
`and glycerol phenylbutyrate, respectively. There was no statistical difference in maximal
`PAA levels between UCD patients who did or did not report neurological AEs. The bottom
`panel depicts mean PAA concentrations (mean [SD]= 61.4 [75.3] vs. 36.4 [55.6]; p = 0.77)
`among patients with cirrhosis and hepatic encephalopathy randomized to treatment with
`glycerol phenylbutyrate who reported neurological adverse events. The range of PAA con-
`centrations as reflected by the box (25th to 75th percentile) are similar for patients who
`did or did not report an AE. The dots depict individual values (See Table 2 for statistical
`summary).
`
`Fig. 2. PAA levels in healthy adults reporting a nervous system adverse event (AE) grouped
`by dose. The maximum PAA value (Cmax) is displayed in relation to dose of glycerol
`phenylbutyrate for patients who did or did not report a neurological adverse event (AE),
`regardless of relationship to study drug or timing relative to blood draw for PAA. The
`box and whisker plots depict mean (horizontal line), 25–75 percentiles (box) and 10
`and 90% confidence intervals. Note that a wide range of PAA levels was observed at each
`dose and among patients with or without AEs. PAA levels were significantly higher
`among patients with AEs as compared to those without at the 6 mL TID dose, but not at
`the 4 mL TID dose. All but 1 subject in the 9 and 12 mL dose groups reported a neurological
`AE (See Table 2 for statistical summary).
`
`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 gener-
`ally mild, started early in the dosing period, and disappeared with
`continued dosing.
`The theoretical risk of PAA toxicity is expected to be similar for sodi-
`um 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 con-
`tinued 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 conver-
`sion 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 the 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 inter-
`esting in this regard that the upward inflection 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 com-
`promised by the fact that concentrations vary considerably over the
`
`[6.1.2.9] [Mokhtarani et al - PAA and PAA-PAGN ratio on line publication.pdf] [Page 5 of 8]
`
`5 of 8
`
`

`

`M. Mokhtarani et al. / Molecular Genetics and Metabolism 110 (2013) 446–453
`
`451
`
`Fig. 3. Plasma PAA intra-subject variability. Healthy subjects and patients with UCD or HE underwent serial blood sampling over 12 to 24 h. The figure depicts the coefficient of variation
`(CV%) as an indicator of intra-subject variability. Regardless of the dose or population, there is high degree of variability among all subjects.
`
`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,
`the plasma PAA fluctuation index varied from 843%–3931%; and fasting
`and maximal PAA levels in HE patients ranged from 0 to 1.3 μg/mL and
`248 to 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 comparatively constant over the day and, therefore, is
`more readily interpretable in a random blood draw.
`These analyses have several limitations. First, although pharmacoki-
`netic 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
`
`Fig. 4. Plasma PAA vs. plasma PAA:PAGN ratio. PAA levels in μg/mL (Y axis) are plotted in
`relation to the ratio of PAA to PAGN concentration (both expressed as μg/mL) in plasma (X
`axis) in that same sample. This plot includes N3500 samples from all populations, includ-
`ing healthy adults (Healthy), patients with cirrhosis and hepatic encephalopathy (Hepat-
`ic), and patients with urea cycle disorders (UCD). All populations exhibit a similar
`relationship, with the upward inflection point occurring at ratios exceeding approximately
`2.5 and PAA concentrations in the range of 100–200 μg/mL.
`
`utilizing the highest recorded PAA for that dosing period. Finally, these
`conclusions pertaining to the absence of a

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