throbber
Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 1 of 18 PageID: 592
`
`John E. Flaherty
`Ravin R. Patel
`McCARTER & ENGLISH LLP
`Four Gateway Center
`100 Mulberry St.
`Newark, NJ 07102
`Telephone: (973) 622-4444
`
`Attorneys for Plaintiff Horizon
`Therapeutics LLC
`
`Robert F. Green
`Caryn C. Borg-Breen
`Ann K. Kotze
`Rachel C. Bell
`GREEN, GRIFFITH & BORG-BREEN LLP
`City Place
`676 N. Michigan Avenue, Suite 3900
`Chicago, Illinois 60611
`Telephone: (312) 883-8000
`
`Of Counsel for Plaintiff Horizon Therapeutics
`LLC
`
`IN THE UNITED STATES DISTRICT COURT
`FOR THE DISTRICT OF NEW JERSEY
`
`HORIZON THERAPEUTICS LLC,
`
`Plaintiff,
`
`v.
`
`Case No. 2:17-cv-5901-KM-MAH
`
`PAR PHARMACEUTICAL, INC.,
`
`Motion Date: See Dkt. No. 57
`
` Defendant.
`
`HIGHLY CONFIDENTIAL –
`PURSUANT TO THE DISCOVERY
`CONFIDENTIALITY ORDER
`
`DECLARATION OF INVENTOR BRUCE F. SCHARSCHMIDT, M.D., IN SUPPORT
`OF PLAINTIFF HORIZON’S OPPOSITION TO PAR’S MOTION FOR SUMMARY
`JUDGMENT OF INVALIDITY UNDER 35 U.S.C. § 101
`
`Horizon Exhibit 2020
`Lupin v. Horizon
`IPR2018-00459
`
`1 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 2 of 18 PageID: 593
`HIGHLY CONFIDENTIAL – PURSUANT TO
`
`TABLE OF CONTENTS
`
`I.
`
`II.
`
`III.
`
`IV.
`
`V.
`
`VI.
`
`INTRODUCTION .............................................................................................................. 1
`
`EDUCATION AND CAREER ........................................................................................... 1
`
`UREA CYCLE DISORDERS ............................................................................................ 2
`
`DEVELOPMENT OF RAVICTI® ..................................................................................... 6
`
`URINARY PAGN OUTPUT .............................................................................................. 8
`
`BLOOD AMMONIA LEVELS .......................................................................................... 9
`
`VII.
`
`THE PAA:PAGN RATIO................................................................................................. 11
`
`i
`
`2 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 3 of 18 PageID: 594
`
`I.
`
`INTRODUCTION
`
`1.
`
`I, Bruce F. Scharschmidt, M.D., submit this declaration on behalf of Horizon
`
`Therapeutics, LLC (“Horizon Therapeutics”) in support of its Opposition to Par’s Motion for
`
`Summary Judgment of Invalidity Under 35 U.S.C. § 101.
`
`2.
`
`I currently serve as a consultant to Horizon Pharma, Inc. (“Horizon Pharma”).
`
`Horizon Therapeutics, the plaintiff in this case, is a subsidiary of Horizon Pharma.
`
`3.
`
`I am one of the named inventors of U.S. Patent No. 9,561,1971 (“the ’197
`
`Patent”), entitled “Methods of Therapeutic Monitoring of Phenylacetic Acid Prodrugs” and
`
`issued on February 7, 2017.
`
`II.
`
`EDUCATION AND CAREER
`
`4.
`
`I completed my undergraduate and medical studies as part of Northwestern
`
`University’s Honors Program in Medical Education, an accelerated program that combined the
`
`traditional four years of undergraduate education and four years of medical education into a six-
`
`year degree. I graduated with my M.D. in 1970.
`
`5.
`
`6.
`
`I am Board Certified in Internal Medicine and Gastroenterology.
`
`After completing my residency and fellowship at the University of California, San
`
`Francisco, I continued my career there for the next 19 years, eventually serving as the Chief of
`
`Gastroenterology and a Professor of Medicine. During this time I also served as Editor-in-Chief
`
`of the Journal of Clinical Investigation and was elected President of the American Society for
`
`Clinical Investigation.
`
`7.
`
`I spent the next ten years at Chiron Corporation, where, as Vice President of
`
`Clinical Development, I headed the clinical development, clinical operations and biostatistics
`
`1 Attached to the Declaration of Rachel C. Bell, Esq. (“Bell Declaration” or “Bell Decl.”),
`submitted contemporaneously, as Ex. A.
`
`1
`
`3 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 4 of 18 PageID: 595
`
`and clinical data management groups before being elevated to the corporate group as VP of
`
`Scientific Affairs. Chiron was acquired by Novartis International AG in April 2006. I continued
`
`working at Novartis as a Vice President until April 2008.
`
`8.
`
`I served as the Chief Medical Officer and subsequently Chief Medical &
`
`Development Officer of Hyperion Therapeutics, Inc. (“Hyperion”) from April 2008 to May
`
`2015, at which time it was acquired by Horizon Pharma. I also served as a Senior Vice President
`
`at Hyperion.
`
`9.
`
`I was listed as an inventor on every patent application filed by Hyperion during
`
`my tenure there.
`
`10.
`
`I have authored over 200 scientific articles over the course of my career.
`
`III.
`
`UREA CYCLE DISORDERS
`
`11.
`
`The ’197 Patent generally is directed to an improved method of treating a patient
`
`with a urea cycle disorder (“UCD”). The method involves measuring the ratio of plasma
`
`phenylacetate (“PAA”) to plasma phenylacetylglutamine (“PAGN”) in the same blood sample of
`
`a UCD patient who has previously taken glycerol phenylbutyrate or another PAA prodrug, and,
`
`if the ratio is outside of the range from 1:1 to 2:1 (or, alternatively, outside of the range from 1:1
`
`to 2.5:1) and, in particular, if the PAA to PAGN ratio is above 2:1 or 2.5:1, administering to the
`
`UCD patient glycerol phenylbutyrate in an amount that is effective to cause the UCD patient to
`
`achieve a PAA to PAGN ratio that is within the target range.
`
`12.
`
`UCDs are a class of inherited metabolic diseases characterized by a partial or
`
`complete absence of one or more enzymes or transporters involved in the metabolic pathway for
`
`disposing of waste nitrogen from the human body in the form of urea, which is excreted in urine.
`
`Disruption of the urea cycle leads to an accumulation of waste nitrogen and a corresponding
`
`increase in ammonia in a patient’s bloodstream. Ammonia is a potent neurotoxin which, when
`
`2
`
`4 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 5 of 18 PageID: 596
`
`present in sufficiently high concentrations in the bloodstream, results in a clinical condition
`
`referred to as hyperammonemia. Hyperammonemia manifests as central nervous system
`
`dysfunction of varying severity up to and including irreversible neurological damage and death if
`
`left untreated. Symptoms of neurotoxicity associated with UCDs include the following:
`
`somnolence (sleepiness), fatigue, lightheadedness, headache, irritability, poor feeding,
`
`hyperventilation, vomiting, disorientation, impaired memory, coma, irreversible neurological
`
`injury and death.
`
`13.
`
`Prior to the advent of drug therapy, patients severely affected by UCDs typically
`
`died. In one longitudinal study, two-thirds of the severely affected babies diagnosed with a UCD
`
`during the first month of life died by the age of 6, even with the administration of sodium
`
`phenylacetate/sodium benzoate 10%/10% rescue treatment (AMMONUL®) during
`
`hyperammonemic crises.2 With the development of newer chronic treatments, survival rates
`
`increased, but one out of five of severely affected newborns diagnosed with a urea cycle disorder
`
`within the first month of life still died from the condition within the first year of life.3
`
`14.
`
`Dietary restriction is a key component of managing treatment of a UCD patient.
`
`Physicians decrease a patient’s protein intake, thereby reducing the amount of waste nitrogen
`
`which must be cleared from the body through a genetically impaired urea cycle. With less waste
`
`nitrogen, patients are less prone to develop elevated blood ammonia levels. However, severe
`
`dietary protein restriction may also decrease the intake of essential amino acids below the levels
`
`2 Marshall Summar et al., Diagnosis, symptoms, frequency and mortality of 260 patients with
`urea cycle disorders from a 21-year, multicenter study of acute hyperammonaemic episodes, 97
`ACTA PAEDIATRICA 1420, 1423 & Fig. 3 (2008), attached as Exhibit B to the Bell Declaration.
`3 See Food & Drug Administration Division Director’s Summary Review of New Drug
`Application No. 203284 (the RAVICTI® (glycerol phenylbutyrate) NDA) at p. 3 of 33, attached
`as Exhibit C to the Bell Declaration.
`
`3
`
`5 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 6 of 18 PageID: 597
`
`necessary for normal growth and development, such that supplementation with amino acid
`
`mixtures or the concomitant use of nitrogen scavenging drugs is often necessary to adequately
`
`balance a UCD patient’s nutritional needs with good metabolic control. In severely affected
`
`patients, dietary management is typically not enough by itself and patients require drug therapy
`
`or other measures. More specifically, in addition to dietary protein restriction, treatment methods
`
`include ammonia-scavenging drugs (alternatively known as nitrogen-scavenging drugs or
`
`alternate pathway drugs) and, in severely affected patients who cannot be controlled by dietary
`
`management and nitrogen scavenging drugs, liver transplantation.
`
`15.
`
`Ammonia-scavenging drugs use a metabolic pathway other than the urea cycle to
`
`excrete excess nitrogen; hence the term alternate pathway drugs. Glycerol phenylbutyrate (GPB),
`
`for example, is a prodrug of PAA. A prodrug is an inactive precursor chemical compound of a
`
`drug, which is then converted to an active form through normal metabolic processes. Glycerol
`
`phenylbutyrate is hydrolyzed by pancreatic enzymes to yield phenylbutyrate (a prodrug of PAA),
`
`which undergoes beta oxidation by the fatty acid oxidation cycle to produce PAA. PAA, in turn,
`
`is converted to PAGN by an enzymatic reaction that conjugates PAA to the amino acid
`
`glutamine. PAGN incorporates two nitrogens and is excreted in the urine, in effect bypassing the
`
`urea cycle.
`
`16.
`
`Alternative pathway drug treatments have increased the survival rate for UCD
`
`patients but are not a cure. Patients with UCDs require daily treatment for life and are managed
`
`by a physician specializing in the disorder. Patient monitoring and treatment compliance is
`
`critically important in ensuring effective treatment of urea cycle disorders and preventing
`
`episodes of acute hyperammonemia.
`
`4
`
`6 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 7 of 18 PageID: 598
`
`17.
`
`Oral administration of drug treatment is widely considered preferable for daily
`
`maintenance of chronic illnesses like urea cycle disorders, as opposed to intravenous or
`
`parenteral administration. However, oral administration of drugs in whatever form – powders,
`
`tablets, capsules, or liquids – requires investigators to consider the taste, odor, and texture of a
`
`drug product during pharmaceutical development. These attributes are collectively referred to as
`
`the “palatability” of a drug product.
`
`18.
`
`Patient compliance is particularly challenging in treatment of pediatric UCD
`
`patients. Generally speaking, pediatric patients are especially sensitive to unpalatable drugs.4
`
`19.
`
`One of the earlier developed ammonia scavenging drugs is BUPHENYL®.5
`
`BUPHENYL® is a sodium phenylbutyrate drug product available in tablet or powder form for
`
`the long-term management of hyperammonemia caused by urea cycle disorders that cannot be
`
`managed by diet alone.6 Unfortunately, the sodium phenylbutyrate in BUPHENYL® is
`
`associated with palatability issues, thereby resulting in reduced patient compliance with
`
`treatment regimens. In one recent study7, 64% of patients reported that sodium phenylbutyrate is
`
`“difficult to take because of its taste and strong odor.” Similarly, nearly half of UCD patients and
`
`their caregivers who stopped sodium phenylbutyrate treatment reported that sodium
`
`4 See Bell Decl. Ex. C p. 29 of 33 (“[T]he product was being developed as [sic] more palatable
`formulation of phenylbutyrate, which would make the prescriber want to use it in young
`children.”).
`5 Hyperion acquired rights to BUPHENYL® in 2013.
`6 See FDA-approved BUPHENYL® Label, attached as Exhibit D to the Bell Declaration.
`7Oleg A. Shchelochkov et al., Barriers to drug adherence in the treatment of urea cycle
`disorders: Assessment of patient, caregiver and provider perspectives, MOLECULAR GENETICS &
`METABOLISM REPORTS 43 (2016), attached as Exhibit E to the Bell Declaration. I am listed as an
`author of this article.
`
`5
`
`7 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 8 of 18 PageID: 599
`
`phenylbutyrate’s taste was a factor contributing to their discontinuation of that course of
`
`treatment.8
`
`20.
`
`Consequently, Hyperion turned its attention to the development of glycerol
`
`phenylbutyrate, hoping to make available to patients a drug product that was biochemically
`
`similar to sodium phenylbutyrate while lacking its undesirable attributes including poor
`
`palatability and high sodium content.9
`
`IV.
`
`DEVELOPMENT OF RAVICTI®
`
`21.
`
`Upon joining Hyperion in 2008, I began working on the clinical trials comparing
`
`glycerol phenylbutyrate oral liquid with BUPHENYL®. The results of these clinical trials were
`
`submitted to the U.S. Food & Drug Administration (“FDA”) as part of Hyperion’s New Drug
`
`Application No. 203284 (“the RAVICTI® NDA”). Ultimately, Hyperion submitted clinical data
`
`from approximately 9 clinical studies for the FDA’s consideration. While the primary purpose of
`
`these clinical trials was to support the RAVICTI® NDA, I also recognized that we had acquired
`
`a large and unique data set and was therefore also interested in whether we could interrogate
`
`these clinical data so as to discover dosing biomarkers that would enable us to provide
`
`physicians with guidelines that could improve methods of treatment and dosage adjustment for
`
`their urea cycle disorder patients undergoing PAA prodrug treatment. In addition to developing a
`
`better drug product, my goal was to develop better guidelines for dose adjustment and
`
`monitoring that could improve outcomes for urea cycle disorder patients.
`
`22.
`
`At the time we were conducting the clinical trials, physicians did not have a
`
`reliable methodology available to help them evaluate the optimal dose of a PAA prodrug drug
`
`8 Id.
`9 U.S. Patent Appl. No. 2010/0008859 (“the ’859 Publication”) at ¶ [0065] (“[HPN-100] avoids
`the unpleasant taste associated with sodium PBA, and it reduces potentially harmful sodium
`intake . . . .”), attached as Exhibit F to the Bell Declaration.
`
`6
`
`8 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 9 of 18 PageID: 600
`
`product for an individual UCD patient. The available scientific literature provided little guidance
`
`to physicians trying to determine the most effective dose and/or adjust the dose to better suit the
`
`needs of a UCD patient. For example, the BUPHENYL® Label contained no guidance on how
`
`to calculate an effective dosage for an individual UCD patient other than including a
`
`recommended range of dosages based on body weight (e.g. 450-600 mg/kg/day) and/or body
`
`surface area of the patient.10 Furthermore, the BUPHENYL® Label provided no guidance on
`
`how to adjust the dosage within that range so as to minimize the risk of toxicity and optimize
`
`control of blood ammonia.11
`
`23.
`
`Because UCDs represent a collection of inherited disorders involving genes
`
`encoding for 6 different enzymes or transporters, and because each of these genes may be
`
`affected by as many as 40 or more different mutations, the UCD patient population is extremely
`
`heterogenous and each patient requires highly individualized treatment. Different UCD patients,
`
`even patients with the same defective gene, may have diseases of varying severity and require
`
`different treatment regimens, including different doses of the same drug. The pediatric
`
`population of UCD patients, particularly infants, tends to include patients with earlier onset and
`
`often more severe disease as well as slower conversion of PAA to PAGN. 12 As a result,
`
`individualized treatment is particularly important in very young UCD patients.
`
`24.
`
`Similarly, because UCDs impair a patient’s ability to excrete waste nitrogen
`
`resulting from break down of dietary protein, the manifestation of a patient’s disease also
`
`10 See Bell Decl. Ex. D at p. 5 of 8, § DOSAGE AND ADMINISTRATION; see also Bell Decl.
`Ex. C at p. 15 of 33 (“[T]he Buphenyl label is not precise in recommending an initial dose . . .
`.”).
`11 See Bell Decl. Ex. D. at p. 5 of 8, § DOSAGE AND ADMINISTRATION.
`12 Bell Decl. Ex. C at p. 15 of 33 (“[I]n children over the age of 2 years, there was higher
`variability and higher concentrations of PAA than in adults.”).
`
`7
`
`9 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 10 of 18 PageID: 601
`
`depends on their diet. Much like diabetes, this means that the manifestation of a UCD patient’s
`
`disease may change from day to day, and even throughout the day, as a result of diet or a
`
`concurrent illness such as an infection.13
`
`25.
`
`Thus, as in to diabetes, I hoped to discover dosing biomarker(s) that would allow
`
`physicians to more effectively tailor the dosing of a phenylbutyrate drug product to the needs of
`
`an individual patient.14 A biomarker is defined as “a characteristic that is objectively measured
`
`and evaluated as an indicator of normal biologic processes, pathogenic processes, or
`
`pharmacologic responses to a therapeutic intervention.”15
`
`26.
`
`Therefore, my colleagues and I undertook extensive analyses of Hyperion’s
`
`uniquely large data set derived from controlled clinical trials to see if it was possible to identify
`
`biomarkers that would help physicians do a better job of treating urea cycle disorder patients.
`
`V.
`
`URINARY PAGN OUTPUT
`
`27.
`
`Initially, our analyses examined the possibility that we could evaluate the efficacy
`
`of glycerol phenylbutyrate by measuring the amount of PAGN excreted over 24 hours in a
`
`patient’s urine. Since PAGN excretes waste nitrogen in place of urea, PAGN does in effect
`
`function as a biomarker of drug effect.
`
`28.
`
`The consensus in the relevant scientific community at this time was that PAA was
`
`completely or almost completely converted to urinary PAGN in UCD patients.16 Through
`
`13 See U.S. Patent No. 9,095,559 at col. 4 ll. 29-64, attached as Exhibit G to the Bell Declaration.
`14 For example, in the diabetes context, HbA1c is an industry-standard biomarker for evaluating
`the efficacy of new drugs and treatments. By contrast, urea cycle disorders have no such agreed-
`upon biomarker by which to evaluate the efficacy of new or existing drugs.
`15 Biomarkers Definitions Working Group, Biomarkers and surrogate endpoints: preferred
`definitions and conceptual framework, 69 CLINICAL PHARMACOLOGY THERAPY 89 (2001)
`attached as Exhibit H to the Bell Declaration.
`16 See, e.g., Brusilow, Phenylacetylglutamine May Replace Urea as a Vehicle for Waste Nitrogen
`Excretion, 29 Pediatric Research, 147-150 (1991) attached as Exhibit I to the Bell Declaration.
`
`8
`
`10 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 11 of 18 PageID: 602
`
`analyzing the clinical data we had collected to support the RAVICTI® NDA, we discovered that
`
`contrary to this consensus, only approximately 60% of the PAA prodrug is converted to urinary
`
`PAGN. Discovering this incomplete and variable conversion rate allowed us to develop more
`
`accurate dosing methods for physicians using PAA prodrugs to treat UCD patients and had
`
`critical implications for UCD treatment.
`
`29.
`
`This research resulted in the issuance of U.S. Patent No. 8,642,012, entitled
`
`“Methods of Treatment Using Ammonia-Scavenging Drugs.”
`
`30.
`
`However, several practical considerations stemming from the need for timed urine
`
`collections are inherent to this dosing approach. In adults, timed 24-hour urine collections are a
`
`nuisance and often incomplete. In infants, timed urine collected requires bladder catheterization,
`
`which is an invasive procedure. Thus, we further analyzed the clinical trial data to develop a
`
`method whereby PAGN concentration in urine could be used to guide dosing of glycerol
`
`phenylbutyrate. Moreover, I continued to analyze the data from Hyperion’s clinical trials in my
`
`effort to further optimize UCD treatment methods.
`
`VI.
`
`BLOOD AMMONIA LEVELS
`
`31.
`
`Next, I considered whether measuring the concentration of ammonia in a patient’s
`
`blood was an effective way to evaluate whether that patient was receiving an effective dosage of
`
`a PAA prodrug. Many of the expert physicians with whom we worked in the clinical trials did
`
`not routinely check blood ammonia, in part because of the inherent variability in ammonia
`
`measurements taken at random times. Moreover, we learned from our studies that blood
`
`ammonia values often varied several-fold throughout the day, even among the well-controlled
`
`individual UCD patients enrolled in our trials. Specifically, we observed that blood ammonia
`
`tended to be lowest in the morning before breakfast and tended to peak in the late afternoon or
`
`9
`
`11 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 12 of 18 PageID: 603
`
`early evening. These findings raised the question of exactly when blood ammonia should be
`
`drawn in UCD patients.
`
`32.
`
`In the Hyperion clinical trials, blood samples were frequently drawn “around the
`
`clock,” i.e., at multiple times during the day and night over 24 hours in order to assess daily
`
`ammonia exposure or burden. In routine clinical practice, such “around the clock” blood draws
`
`are impractical and rarely performed. Thus, we had at our disposal a unique data set that
`
`permitted us to gain insights never before appreciated.
`
`33. We discovered through extensive statistical analyses of the clinical data that there
`
`is a strong correlation between a patient’s fasting plasma ammonia level and that patient’s
`
`overall daily ammonia exposure. This was an important finding. It meant “around the clock”
`
`blood draws are not needed to assess daily ammonia exposure and average blood ammonia
`
`levels. Instead, physicians can assess daily blood ammonia exposure based on fasting blood
`
`ammonia, i.e., the first blood draw of the morning before patients eat breakfast or take their first
`
`daily dose of any drugs.
`
`34.
`
`Using that correlation, we next discovered that an individual patient’s likelihood
`
`of experiencing a normal daily ammonia exposure is enhanced if drug therapy is adjusted so as to
`
`maintain a fasting ammonia value that does not exceed half the upper limit of normal (“ULN”).
`
`We further discovered that keeping fasting ammonia levels to within half the upper limits of
`
`normal decreases both the risk and frequency of hyperammonemic crises.17 These discoveries
`
`allow us to develop a methodology whereby physicians could adjust the dose of a nitrogen-
`
`17 Lee et al., Blood ammonia and glutamine as predicators of hyperammonemic crises in patients
`with urea cycle disorder, Genetics in Medicine, 17(7):561-568 (2015) at 561, 563-565, attached
`as Exhibit J to the Bell Declaration.
`
`10
`
`12 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 13 of 18 PageID: 604
`
`scavenging drug based on measuring a fasting blood ammonia level and comparing it to the
`
`ULN, where a fasting level that is greater than half the ULN requires an increased dosage.
`
`35. We filed two provisional patent applications disclosing our method of adjusting
`
`treatment of a patient with a urea cycle disorder based on that patient’s blood ammonia level
`
`from a fasting blood draw in the fall of 2011. These applications eventually led to U.S. Patent
`
`Nos. 8,404,215; 9,095,55918; 9,254,278; and 9,326,966.
`
`36.
`
`However, although we had developed the above improved methods of
`
`determining an effective dose of a PAA prodrug to treat UCD patients, we were still concerned
`
`about the potential risk of neurotoxicity due to elevated levels of PAA that might result from too
`
`high a dosage of phenylbutyrate. This concern was based on reports by Thibault et al. in the
`
`context of cancer patients that reversible neurotoxicity was associated with plasma PAA levels
`
`exceeding ~500 µmol/L.19 We continued to examine the clinical data in the hopes of locating a
`
`biomarker that would indicate the optimal dose of phenylbutyrate to a physician treating a UCD
`
`patient.
`
`VII. THE PAA:PAGN RATIO
`
`37. We decided to look at the relative concentrations of PAA and PAGN in blood.
`
`38.
`
`PAA and PAGN have comparatively short circulating half-lives. Therefore,
`
`because PAA and PAGN levels in a patient’s blood vary many-fold over the course of the day
`
`depending on when the sample is drawn in relation to drug dosing, random blood PAA values as
`
`might be drawn during a routine clinic visit are of limited utility in determining whether that
`
`18 See, e.g., Bell Decl. Ex. G.
`19 See, e.g., Alain Thibault et al., Phase I Study of Phenylacetate Administered Twice Daily to
`Patients with Cancer, 75 CANCER 2932, 2937 (1995), attached as Exhibit K to the Bell
`Declaration; see also Bell Decl. Ex. D at p. 4 of 8, § CLINICAL ADVERSE EVENTS.
`
`11
`
`13 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 14 of 18 PageID: 605
`
`patient might, at some point during the day, experience levels sufficiently high as to be
`
`associated with neurologic adverse events. Consequently, clinicians did not consider a patient’s
`
`blood PAA or PAGN levels to be useful in determining whether a patient was receiving the
`
`correct dose of a phenylbutyrate drug.20
`
`39.
`
`Although it had previously been established that PAGN was a metabolite of PAA,
`
`the relationship between these two metabolites in relation to drug dosing was not appreciated in
`
`the prior art. The published research at that time provided no indication that the relative
`
`concentrations of PAA and PAGN could provide useful clinical information of any type, let
`
`alone provide definitive dosing guidelines regarding a patient’s risk of neurotoxicity.
`
`40.
`
`Based on our analysis of the clinical data, we theorized that the enzymatic
`
`conversion of PAA to PAGN, which is rate-limiting in overall metabolism, might at some point
`
`become saturated. We further theorized, based on the Michaelis-Menten model of enzyme
`
`kinetics which describes the reaction process in which an enzyme converts a substrate into a
`
`product, that an elevated ratio of precursor (i.e., PAA) to substrate (i.e., PAGN) might serve as a
`
`biomarker indicating that PAA to PAGN conversion was saturated or approaching saturation;
`
`20 See, e.g., Uta Lichter-Konecki et al., Ammonia Control in Children with Urea Cycle Disorders
`(UCDs): Phase 2 Comparison of Sodium Phenylbutyrate and Glycerol Phenylbutyrate, 103
`Molecular Genetics and Metabolism 323, at 328 Table 3 (2011) (“[B]lood levels of PBA, PAA
`and PAGN all varied widely over the course of the day. . . .”), attached as Exhibit L to the Bell
`Declaration; Brendan Lee et al., Phase 2 Comparison of a Novel Ammonia Scavenging Agent
`with Sodium Phenylbutyrate in Patients with Urea Cycle Disorders: Safety, Pharmacokinetics
`and Ammonia Control, 100 Molecular Genetics & Metabolism 221, at 226 Table 3 (2010),
`attached as Exhibit M to the Bell Declaration; U.S. Patent Appl. No. 2012/0022157 at [004]
`(“[M]easuring the blood level of the prodrug (e.g., PBA) or of PAA formed from it is unreliable
`in assessing drug effect . . . .”), attached as Exhibit N to the Bell Declaration; Bell Decl. Ex. F at
`[0023], [0070], & [0072] (“[S]ystemic levels of PAA or PBA are not reliably correlated with the
`efficacy of HPN-100 as an ammonia scavenger.”); see also Bell Decl. Ex. A col. 10 ll. 4-7
`(“Since PAA, PAGN, and ammonia levels do not provide the information necessary to determine
`whether a subject is effectively converting PBA to PAGN (i.e., effectively utilizing the PAA
`prodrug) . . . .”).
`
`12
`
`14 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 15 of 18 PageID: 606
`
`i.e., had reached its maximum level. For a patient whose PAA to PAGN conversion was
`
`approaching saturation, prescribing additional phenylacetate prodrug (i.e., glycerol
`
`phenylbutyrate or sodium phenylbutyrate) would not result in further formation of
`
`phenylacetylglutamine and thereby removal of waste nitrogen, but could result instead in
`
`elevation of circulating PAA levels that could put the patient at risk of toxicity.
`
`41. We believed that if an elevated ratio of PAA to PAGN was a biomarker of
`
`saturation, then we might be able to identify a specific value of the PAA:PAGN ratio that would
`
`tell us when the enzymatic reaction was saturated. More specifically, we hoped we could identify
`
`a ratio of PAA:PAGN above which the risk of phenylacetate toxicity was substantially increased
`
`and develop a biomarker more useful than the highly variable individual levels of PAA and
`
`PAGN.
`
`42.
`
`Our extensive review of Hyperion’s clinical data revealed, to our surprise, that
`
`while the individual concentrations of PAA and PAGN fluctuated widely throughout the day, a
`
`patient’s ratio of PAA:PAGN remained comparatively constant.
`
`43. We were also surprised to find that the utility of the ratio of PAA:PAGN blood
`
`levels was largely consistent among individual patients as well as patient populations, despite the
`
`heterogeneous nature of the UCD patient population. As we explained in the ’197 patent: 21
`
`The excess of PAA over PAGN, indicated by larger ratios as PAA increases, was
`evident across all dosage groups, disease populations, and types of treatment in
`UCD patients (i.e., applies to both NaPBA and HPN-100). This finding suggests
`that analysis of the precursor (PAA) to product (PAGN) ratio may be predictive
`of the efficiency of conversion among patients with or without liver
`dysfunction . . . and independently of dose.
`
`21 Bell Decl. Ex. A at col. 24 ll. 22-30.
`
`13
`
`15 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 16 of 18 PageID: 607
`
`44.
`
`Ultimately, we concluded that a ratio of PAA:PAGN above 2:1 in a patient’s
`
`fasting blood draw indicated that the metabolic reaction converting phenylacetate to
`
`phenylacetylglutamine may have become saturated. We confirmed through statistical analysis
`
`that patients with a ratio of PAA:PAGN greater than 2:1 (or, alternatively, greater than 2.5:1)
`
`were at a dramatically higher risk of experiencing neurotoxic side effects from receiving too high
`
`of a dose of glycerol phenylbutyrate. 22 Consequently, a ratio of PAA:PAGN above 2:1 informed
`
`the treating physician that the patient’s dose of glycerol phenylbutyrate needed to be reduced, or,
`
`if that patient was also experiencing elevated blood ammonia levels which made dose reduction
`
`problematic, alteration of dosing schedule.
`
`45.
`
`Similarly, we concluded that a ratio of PAA:PAGN below 1:1 in a patient’s
`
`fasting blood draw could indicate that the dosage of nitrogen scavenging medication was
`
`potentially insufficient. Consequently, a ratio of PAA:PAGN below 1:1 suggests to the treating
`
`physician that the UCD patient’s dose of glycerol phenylbutyrate may need to be increased, and
`
`that such increase could be safely made, if also warranted by the subject’s clinical status
`
`including ammonia level, with minimal risk of triggering high PAA levels.23
`
`46.
`
`In the Hyperion clinical trials, just as was the case for ammonia, PAA and PAGN
`
`levels were drawn ‘around the clock’ in order to assess maximum PAA levels. In clinical
`
`practice, ‘around the clock’ blood draws are impractical and, as described above, random PAA
`
`blood draws as might be done at a routine visit, are of little utility. By contrast, the comparative
`
`constancy of the PAA:PAGN ratio renders it useful in the context of routine clinical practice for
`
`22 Id. at col. 27 ll. 38-40 (“Furthermore, ratios between 2.0-2.5 were associated with significantly
`higher PAA levels than ratios less than 2.0 (p<0.001).”)
`23 Id. at col. 18 ll. 39-43 (“In certain of these embodiments where the target range is 1 to 2.5, a
`PAA:PAGN ratio below 1 indicates the PAA prodrug dosage is unlikely to be effective and
`needs to be adjusted upwards.”).
`
`14
`
`16 of 18
`
`

`

`Case 2:17-cv-05901-KM-MAH Document 62-3 Filed 04/16/18 Page 17 of 18 PageID: 608
`
`purposes of assessing that patient’s risk of PAA toxicity individualizing dosing irrespective of
`
`the specific genetic cause of a patient’s urea cycle disorder. For the first time, there was a
`
`relatively straightforward blood test that allowed a clinician to adjust a p

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