throbber
Vol. 7, 3047–3055, October 2001
`
`Clinical Cancer Research
`
`3047
`
`A Phase I Clinical and Pharmacological Evaluation of Sodium
`Phenylbutyrate on an 120-h Infusion Schedule1
`
`Michael A. Carducci,2 Jill Gilbert,
`M. Katherine Bowling, Dennis Noe,
`Mario A. Eisenberger, Victoria Sinibaldi,
`Yelena Zabelina, Tian-ling Chen,
`Louise B. Grochow, and Ross C. Donehower
`Divisions of Medical Oncology [M. A. C., J. G., M. K. B., M. A. E.,
`V. S., L. B. G., R. C. D.] and Experimental Therapeutics and
`Pharmacology [M. A. C., D. N., Y. Z., T-l. C., L. B. G.], The Johns
`Hopkins Oncology Center, The Johns Hopkins University School of
`Medicine, Baltimore, Maryland 21231
`
`ABSTRACT
`Purpose: Sodium phenylbutyrate (PB) demonstrates
`potent differentiating capacity in multiple hematopoietic
`and solid tumor cell lines. We conducted a Phase I and
`pharmacokinetic study of PB by continuous infusion to
`characterize the maximum tolerated dose, toxicities, phar-
`macokinetics, and antitumor effects in patients with refrac-
`tory solid tumors.
`Patients and Methods: Patients were treated with a
`120-h PB infusion every 21 days. The dose was escalated
`from 150 to 515 mg/kg/day. Pharmacokinetics were per-
`formed during and after the first infusion period using a
`validated high-performance liquid chromatographic assay
`and single compartmental pharmacokinetic model for PB
`and its principal metabolite, phenylacetate.
`Results: A total of 24 patients were enrolled on study,
`with hormone refractory prostate cancer being the predom-
`inant tumor type. All patients were evaluable for toxicity
`and response. A total of 89 cycles were administered. The
`dose-limiting toxicity (DLT) was neuro-cortical, exemplified
`by excessive somnolence and confusion and accompanied by
`clinically significant hypokalemia, hyponatremia, and hy-
`peruricemia. One patient at 515 mg/kg/day and another at
`345 mg/kg/day experienced this DLT. Toxicity resolved <12
`h of discontinuing the infusion. Other toxicities were mild,
`including fatigue and nausea. The maximum tolerated dose
`
`Received 3/14/01; revised 6/11/01; accepted 6/13/01.
`The costs of publication of this article were defrayed in part by the
`payment of page charges. This article must therefore be hereby marked
`advertisement in accordance with 18 U.S.C. Section 1734 solely to
`indicate this fact.
`1 Supported by UO-1 CA 70095, American Society of Clinical Oncol-
`ogy Young Investigator Award; NIH-K08-CA-69164; NIH-R01-
`CA75525 (to M. A. C.); Aegon Scholarship in Oncology (to J. G.),
`General Clinical Research Center N01-CM07302 from the National
`Cancer Institute.
`2 To whom requests for reprints should be addressed, at The Johns
`Hopkins Oncology Center, 1 M88 Bunting-Blaustein Cancer Research
`Building, 1650 Orleans Street, Baltimore, MD 21231-1000. E-mail:
`carducci@jhmi.edu.
`
`was 410 mg/kg/day for 5 days. Pharmacokinetics demon-
`strated that plasma clearance of PB increased in a continu-
`ous fashion beginning 24 h into the infusion. In individuals
`whose Vmax for drug elimination was less than their drug-
`dosing rate, the active metabolite phenylacetate accumu-
`lated progressively. Plasma PB concentrations (at 410 mg/
`kg/day) remained above the targeted therapeutic threshold
`of 500 ␮mol/liter required for in vitro activity.
`Conclusion: The DLT in this Phase I study for infu-
`sional PB given for 5 days every 21 days is neuro-cortical in
`nature. The recommended Phase II dose is 410 mg/kg/day
`for 120 h.
`
`INTRODUCTION
`Differentiation therapy for epithelial malignancies may po-
`tentially alter tumor growth and progression, slow or inhibit
`metastases, inhibit angiogenesis, and/or effect response to other
`forms of therapy (1–5). Nonretinoid agents such as hexameth-
`ylenebisacetamide and sodium butyrate have been evaluated
`clinically, but sustained systemic levels required for activity
`have not been achieved because of toxicity and/or lack of a
`suitable formulation (6 –9). Sodium PB,3 an aromatic fatty acid,
`is a lead candidate as a cancer differentiating agent and a histone
`deacetylase inhibitor (10 –13). Sodium PB is the precursor to
`PA, and both compounds are potent differentiating agents in
`vitro (10 –18). PA is formed by ␤-oxidation of PB (19). PB is
`Food and Drug Administration approved for children and adults
`with hyperammonemia associated with urea cycle disorders
`(recommended dose of 13 g/m2/day) and has been used for adult
`patients with hyperammonemia secondary to high-dose chemo-
`therapy for leukemia and transplant therapies (19 –22). PB is
`also under investigation for cystic fibrosis and adrenal leukodys-
`trophy.
`Preclinical studies have looked at the ability of these drugs
`to alter gene expression and promote differentiation (23–25). In
`various tumor model systems, PA/PB can arrest cells in G1-G0
`with induction of p21WAF1 and other cdk-2-associated cell cycle
`checkpoint proteins, alter expression of invasion products such
`as urokinase-plasminogen activator (UPA), induce apoptosis,
`inhibit telomerase, and increase MHC class I expression at
`concentrations of 500-2500 ␮mol/liter PB (23–31). Delay in
`tumor progression has been noticed in prostate and malignant
`glioma models (12, 16, 17). It should be noted that tumor
`markers, such as PSA, might not be the most accurate measure-
`ments of progressive disease in patients treated with PB, as a
`rise in tumor markers may signal cell differentiation rather than
`
`3 The abbreviations used are: PB, phenylbutyrate; PA, phenylacetate;
`PG, phenylacetylglutamine; PSA, prostate-specific antigen; MTD, max-
`imum tolerated dose; NCI, National Cancer Institute; DLT, dose-limit-
`ing toxicity; PCA, prostate cancer.
`
`
`
`clincancerres.aacrjournals.org Downloaded from
`
`on August 3, 2018. © 2001 American Association for Cancer
`Research.
`
`Par Pharmaceutical, Inc. Ex. 1007
`Par v. Horizon, IPR of Patent No. 9,561,197
`Page 1 of 10
`
`

`

`3048 Phase I Study of Phenylbutyrate
`
`disease progression. Proposed mechanisms of action to elicit
`these effects include modification of lipid metabolism, activa-
`tion of the peroxisome proliferator activator receptor, mainte-
`nance of histone acetylation through inhibition of histone
`deacetylase, inhibition of protein isoprenylation, and/or gluta-
`mine starvation (10, 27–31). There may be other cellular and
`molecular effects of PB that contribute to its antitumor activity.
`In addition to all of the potential cellular and molecular
`effects, PB is minimally toxic in children with urea cycle dis-
`orders and hemoglobinopathies (31). Serum concentrations of
`PB have been reported in the range (500 –2500 ␮mol/liter)
`required for in vitro effects of growth arrest and differentiation.
`Preclinical data from our laboratory and others suggest PB may
`be more potent and produce more molecular effects than PA
`(12). PB has activity similar to butyrate (induction of apoptosis
`and histone acetylation) that PA may not possess. For these
`reasons, Phase I studies of PB were initiated.
`The purpose of this Phase I trial was to determine the MTD
`and toxicity profile of PB administered i.v. as an 120-h infusion
`every 21 days and to describe the pharmacokinetic behavior of
`this novel agent in patients with refractory solid tumors.
`
`PATIENTS AND METHODS
`Patient Eligibility
`The protocol was approved by the Joint Committee on
`Clinical Investigation of the Johns Hopkins University School
`of Medicine. Male and female patients ⱖ18 years of age with a
`histologically or cytologically documented diagnosis of cancer
`(solid tumor or lymphoma) refractory to conventional therapeu-
`tic modalities were eligible. All patients enrolled onto the study
`had progressing, evaluable, or measurable disease. Patients were
`required to have an Eastern Cooperative Oncology Group per-
`formance status of 0, 1, or 2 and a life expectancy of ⱖ3 months.
`Patients were not to have had major surgery, radiotherapy, or
`chemotherapy in ⱕ28 days and were to be fully recovered from
`the toxicity of prior therapy. Patients with prostate cancer were
`allowed to continue on a leutenizing hormone releasing hor-
`mone (LHRH) agonist. A 4 – 6-week period of antiandrogen
`(flutamide and bicalutamide) withdrawal and evidence of PSA
`progression was required of all men on an antiandrogen before
`entry. Patients that previously received suramin were eligible if
`their suramin level was ⬍50 ␮g/ml. Adequate organ function
`was required at study entry: WBC ⬎ 2,000 or absolute neutro-
`phil count (ANC) ⬎ 1,500/mm3, platelets ⬎ 100,000/mm3, and
`hemoglobin ⬎ 9 g/dl, a serum creatinine ⬍ 2 mg/dl, total
`bilirubin ⬍ 1.5, and aspartate aminotransferase/alanine amino-
`transferase ⬍ 1.5 ⫻ the upper limit of normal, left ventricular
`ejection fraction (LVEF) by MUGA or echocardiogram ⬎ 40%
`and no history of congestive heart failure (CHF) requiring
`hospitalization, or uncontrolled hypertension (diastolic blood
`pressure (BP) ⬎110 mmHg) and a forced expiratory volume
`⬎ 1.5 liter/min. Patients with obstructive uropathy were
`(FEV)1
`eligible if obstruction was relieved by nephrostomy or other
`appropriate intervention. Patients were required to give in-
`formed consent with understanding of the investigational nature
`of the treatment and its potential risks. Patients were ineligible
`if they had an active infectious process, including HIV or viral
`hepatitis; a malignant brain tumor, or known central nervous
`
`system metastases even if treated previously and not clinically
`active; an active seizure disorder; or a baseline dementia with
`mini-mental score ⬍ 23 (32). Pregnant or nursing females were
`ineligible. Patients with requirement for steroids for any reason,
`including previous suramin therapy, were excluded because of
`concerns about potential interference with the mechanism of
`action of PB.
`
`Pretreatment Evaluation
`Patients were assessed before receiving the first dose of PB
`with a complete history, including prescription and nonprescrip-
`tion drug history, physical exam with a thorough neurological
`examination, including a mini-mental status examination (32),
`and performance status assessment. Documentation of evaluable
`or measurable disease was performed ⱕ4 weeks of initiation of
`therapy. Pretreatment laboratory evaluations included a com-
`plete blood count with platelet and differential, chemistry panel
`(including electrolytes, creatinine, bilirubin, aspartate amino-
`transferase, alanine aminotransferase, alkaline phosphatase, uric
`acid, total protein, albumin, calcium, phosphorous, and magne-
`sium), partial
`thromboplastin time and prothrombin time,
`urinalysis, human chorionic gonadotropin (if woman of child-
`bearing age), EKG, MUGA or echocardiogram for LVEF,
`spirometry with lung volumes, chest roentgenogram (or chest
`computed tomography if part of the tumor assessment), appro-
`priate tumor marker (i.e., PSA, CA-125, CA15-3, and CA19-9),
`and a suramin level, if the patient had been treated previously
`with suramin.
`
`Evaluation During Therapy
`Patients were monitored with laboratory and clinical eval-
`uations weekly by an investigator or research nurse while on
`study. The history and physical was performed at the beginning
`of each 5-day infusion. Patients were also seen as needed for
`additional toxicity assessment. Toxicity was graded using the
`NCI common toxicity criteria, version 2.0. Tumor markers were
`assessed monthly. Evaluation of measurable and evaluable dis-
`ease was performed after each two cycles of therapy. Documen-
`tation of performance status was required with all visits. Com-
`plete response was defined as the disappearance of all clinical
`evidence of active tumor and symptoms for ⱖ1 month. Partial
`response required a ⬎50% decrease in the sum of the products
`of the perpendicular tumor diameters of all measurable lesions
`for ⱖ4 weeks without simultaneous increase in the size of any
`lesion or appearance of any new lesion. Progressive disease was
`defined as a ⬎25% increase in the size of any measurable lesion
`or the appearance of any new lesion. Stable disease represented
`a response less than a partial response or growth less than
`progressive disease. Changes in tumor markers did not factor in
`to the definition of response, e.g., a rising PSA did not define
`progression. Changes in PSA are merely descriptive.
`
`Treatment Plan
`The Investigational Drug Branch of the NCI received phe-
`nylbutyric acid, sodium salt (National Service Center number
`657802; EL532) from Elan Pharmaceutical Research Corp.,
`Gainesville, GA. PB was formulated in 50-ml glass vials con-
`taining 40% viscous solution of sodium PB in sterile water (400
`
`
`
`Downloaded from on August 3, 2018. © 2001 American Association for Cancerclincancerres.aacrjournals.org
`
`
`Research.
`
`Par Pharmaceutical, Inc. Ex. 1007
`Par v. Horizon, IPR of Patent No. 9,561,197
`Page 2 of 10
`
`

`

`Clinical Cancer Research
`
`3049
`
`mg/ml). The sodium content of one vial was 4.9 grams of
`sodium. PB was provided for the patients on this study by the
`Division of Cancer Treatment, NCI.
`Patients were admitted to the General Clinical Research
`Center of the Johns Hopkins Hospital for each course of therapy.
`PB was administered as a continuous i.v. infusion (centrally or
`peripherally) for 120 h every 21 days. A course was defined as
`the 21-day cycle, consisting of the 5-day infusion and 16 days of
`rest. The infusional starting dose was 150 mg/kg/day for 5 days,
`a dose below that shown previously to be safe in children with
`urea cycle disorders. Subsequent dose levels were 225, 285,
`345, 410, and 515 mg/kg/day for 5 days. Doses could be
`escalated within individual patients if 3 patients at the next
`higher dose level had received therapy for ⬎1 month without
`DLT. Patients remained on study until evidence of progressive
`disease, severe DLT, or patient request to discontinue therapy.
`DLT was defined using the NCI common toxicity criteria, as
`neurotoxicity ⱖ grade 2 or any other toxicity ⱖ grade 3. A
`registered nutritionist met with every patient to discuss salt
`intake and to provide recommendations of lowering sodium
`intake while receiving PB.
`Cohorts of 4 patients were initially enrolled at each dose
`level to fully evaluate the central nervous system toxicity and to
`gather more pharmacokinetic data. If no patients in a cohort
`experienced a DLT during the initial 3-week observation period,
`the dose was escalated, and the next cohort was commenced. If
`1 of 4 patients experienced a DLT, 2 additional patients were
`added at that dose level. If 2 patients experienced a DLT at any
`dose level, no additional patients were enrolled at that dose
`level, and an additional 2 patients were entered at the next lower
`dose level. Dose escalation could occur once all 4 patients in a
`given cohort were treated, and at least 2 patients had completed
`one course (21 days) with the 3rd and 4th patients ⱖ1 week into
`therapy without meeting criteria for the MTD. Although the
`study was not designed to escalate doses within patients, in-
`trapatient dose escalation was allowed. A patient who received
`at least two courses of treatment without toxicity (except alo-
`pecia) greater than grade II at a given dose level could have their
`subsequent treatments at the next higher dose level. Before such
`an escalation could occur, 2 PB-naı¨ve patients must have com-
`pleted at least one course (22 days) at the next higher dose level
`without any DLT. The MTD was defined as that dose level at
`which consistent, definable toxicity occurs that is reversible and
`does not subject patients to excessive risk or discomfort.
`Patients who received ⱖ1 day of PB were assessable for
`toxicity, but only those patients who received one full 21-day
`course of PB were assessable for response. Patients for whom a
`drug was held because of DLT still had to complete the full
`21-day observation period. Any patient who experienced DLT
`and recovered fully from that toxicity could resume PB at the
`next lower dose level; however, for MTD determination, pa-
`tients were included only at their initial, higher dose level.
`Patients who did not recover fully from DLT ⱕ14 days or had
`a DLT at a subsequent lower dose were taken off study. The
`study was defined as complete for a given patient if the patient
`did not recover fully from a DLT ⱕ14 days or had a DLT at a
`subsequent lower dose. Patients were also removed from the
`study if they had evidence of progressive disease or if the patient
`requested to be taken off the study.
`
`Table 1 Gradient profile for HPLC
`
`Time (min)
`0.00
`20.00
`23.00
`35.00
`35.01
`38.00
`38.01
`45.00
`
`A (%)
`47.5
`38.5
`38.5
`27.5
`5.0
`5.0
`47.5
`47.5
`
`B (%)
`5.0
`23.0
`23.0
`45.0
`90.0
`90.0
`5.0
`5.0
`
`C (%)
`47.5
`38.5
`38.5
`27.5
`5.0
`5.0
`47.5
`47.5
`
`Recognizing that the recommended Phase II dose is often
`one dose level below the MTD, the recommended Phase II dose
`was to be dictated by toxicities, patient tolerability, and plasma
`levels achieved. An objective of this study was to achieve a
`range of plasma levels that may be clinically relevant as dictated
`by preclinical models.
`
`Pharmacokinetic Study
`Pharmacokinetic Sampling. Drug disposition studies
`were performed during the first course of treatment. Blood
`specimens were collected before the start of the infusion, at 0.5,
`1, 2, 4, 6, 12, 24, 36, 48, 60, and 96 h during the infusion and
`just before completion of the infusion (120 h). Postinfusion
`specimens were obtained at 0.5, 1, 2, 4, 8, and 24 h as measured
`from the end of the infusion. Urine specimens (every 24 h) were
`collected between 0 and 24 h and between 96 and 120 h as
`measured from the start of the infusion.
`Analytic Assay. Plasma PB, PA, and PG concentrations
`were determined in all blood specimens by reverse-phase high-
`performance liquid chromatography assay. These methods have
`not been published previously. Plasma (200 ␮l) containing
`compound was transferred to a microcentrifuge tube, followed
`by the addition of 50 ␮l of 10% perchloric acid (Sigma Chem-
`ical Co., St. Louis, MO) for protein extraction. The samples
`were vortexed and centrifuged (International Equipment Co.,
`Needham Heights, MA) at 4°C at 8500 rpm for 10 min. After
`centrifugation, 150 ␮l of supernatant was added to 5 ␮l of
`super-saturated potassium bicarbonate solution for neutraliza-
`tion and centrifuged again at 4°C at 8500 rpm for 10 min. All of
`the supernatant was transferred to an autosampler vial for anal-
`ysis. The chromatographic apparatus consisted of a Hewlett
`Packard Series II 1090 Liquid Chromatographic (Hewlett Pack-
`ard Corp., Palo Alto, CA) with an autosampler compartment,
`solvent delivery system, and a diode-array UV absorbance de-
`tector with a resolution of 2 nm. The absorbance wavelength
`was 208 nm (bandwidth, 10 nm) with a reference wavelength of
`400 nm (bandwidth, 80 nm). Mobile phase A consisted of 100%
`deionized water (Milli-QUV Plus; Millipore Corp., Bedford,
`MA) with 0.005 M phosphoric acid (Sigma Chemical Co.)
`buffer. Mobile phase B consisted of 100% high-performance
`liquid chromatography grade acetonitrile (J. T. Baker, Phillips-
`burg, NJ) with 0.005 M phosphoric acid buffer, and mobile
`phase C was identical to mobile phase A. All mobile phases
`were run at a flow rate of 1 ml/min for a run time of 45 min at
`the gradient profile listed on Table 1.
`A Waters Nova-Pak C18 guard column was placed in line
`before the analytical column. The samples were injected onto a
`
`
`
`clincancerres.aacrjournals.org Downloaded from
`
`on August 3, 2018. © 2001 American Association for Cancer
`Research.
`
`Par Pharmaceutical, Inc. Ex. 1007
`Par v. Horizon, IPR of Patent No. 9,561,197
`Page 3 of 10
`
`

`

`3050 Phase I Study of Phenylbutyrate
`
`Table 2 Patient characteristics
`
`Total patientsa
`Male
`Female
`Diagnosis
`Prostate, hormone-refractory
`Kidney
`Melanoma
`Endometrium
`Performance status
`0
`1
`2
`Prior therapy
`Chemotherapy, XRT, hormone
`XRT,b hormone
`Hormone
`Chemotherapy, hormone
`Chemotherapy, radiation
`Immunotherapy only
`Chemotherapy, immunotherapy
`No therapy
`a Age: range, 48 – 80, median, 66.
`b XRT, X-ray therapy.
`
`24
`23
`1
`
`19
`3
`1
`1
`
`5
`18
`1
`
`7
`6
`4
`3
`1
`1
`1
`1
`
`lyzed by nonlinear mixed effect modeling. In the pharmacoki-
`netic models used, PA was modeled as arising from PB via the
`Michaelis-Menten-type saturable elimination process for PB
`(Fig. 1). PG was modeled as arising from PA via a Michaelis-
`Menten-type saturable process. For both species, it was assumed
`that biotransformation was complete. PA was modeled as dis-
`tributing in a single compartment with elimination solely via the
`process that gives rise to PG. The Vmax of this elimination
`process was treated as increasing in an exponential fashion
`beginning 24 h after the institution of drug therapy. PG was
`modeled as distributing in a single compartment with first-order
`elimination. In one model, the elimination was treated as time
`invariant, and in another, it was modeled as increasing in an
`exponential fashion beginning 24 h after the institution of drug
`therapy.
`Urine clearance rates of PG were calculated as the amount
`of PG excreted in the urine during the period of urine collection
`divided by the area under the PG plasma disposition curve
`during the same period of time. The areas were determined
`using a linear trapezoidal rule.
`
`RESULTS
`Patients (24) were enrolled at six dose levels. Patient char-
`acteristics are listed in Table 2. A total of 89 full cycles of
`treatment were administered. All patients were evaluable for
`toxicity and response assessment. The majority of patients were
`men with advanced hormone refractory prostate cancer. Over
`half of the patients had received previous chemotherapy. One
`patient with renal cell cancer had had no previous therapy, and
`4 men with prostate cancer received PB as their first therapy in
`the hormone refractory state. The median number of courses per
`patient was four (range, one to eight) for an average time on trial
`of 84 days.
`Patients (21) were withdrawn from therapy secondary to
`progressive disease documented by radiograph or symptoms.
`
`Fig. 1 Single compartment model of PB disposition. PB is ␤-oxidized
`to PA, and glutamine binds PA to form PG, which is cleared in the urine.
`The disposition model for these three compounds is defined by a
`specific Vmax and km for the disposition of one compound to another.
`
`reverse-phase (Waters Bond-Pak C18, 3.9 ⫻ 300 mm; Millipore
`Corp) column, which was maintained at 60°C. PA, PB, and PG
`had retention times of ⬃18.2, 31.4, and 10.2 min, respectively.
`Chromatographic peak area was used for quantitation by linear
`regression analysis. The lower limit of detection for the assay
`was 5 ␮g/ml. Quality control samples were assayed at concen-
`trations of 15, 35, and 85 ␮g/ml, and the inter-day coefficients
`of variation were 11% for PA, ⬍7% for PB, and ⬍11% for PG.
`One set of quality control samples were placed before the
`calibration standards, before the patient samples, and then im-
`mediately after the patient samples were assayed. Samples from
`1 patient were assayed with each analytical run. Each analytical
`run was equivalent to ⬃26 h; autosampler stability for 35 h
`demonstrated that metabolites are stable for this period of time
`with a ⬍10% change (loss or gain) in concentration. The lower
`limit of quantitation was 5 ␮g/ml for PA and PG and 10 ␮g/ml
`for PB. PG concentrations were determined in the urine speci-
`mens, and the amount of PG excreted during the urine collection
`period was calculated as concentration ⫻ urine volume. The
`accuracy of the assay was 93–98% with precision of the assay to
`ⱕ1– 4%.
`Pharmacokinetic Methods. The plasma PB disposition
`curves were analyzed in two ways: (a) individual subject dis-
`position curves were fit by nonlinear regression (PCNONLIN;
`Scientific Consulting, Apex, NC) using a pharmacokinetic
`model consisting of a single distribution compartment and a
`first-order elimination process that increases in magnitude in an
`exponential fashion beginning 24 h after the institution of drug
`therapy, i.e., clearance rate(t ⬎ 24 h) ⫽ clearance rate(t ⫽
`0)⫻exp[kinc⫻(t ⫺ 24 h)], where kinc is the rate constant of the
`exponential process; and (b) the data were analyzed using a
`pharmacokinetic model consisting of a single distribution com-
`partment and a Michaelis-Menten-type saturable elimination
`process in which Vmax increases in magnitude in an exponential
`fashion beginning 24 h after the institution of drug therapy. For
`this model, the disposition curves were analyzed by nonlinear
`mixed effect modeling (P-Pharm; MicroPharm International,
`Champs-sur-Marne, France). With this software, population pa-
`rameter values were estimated using an EM-type iterative algo-
`rithm, and individual subject parameter values were estimated
`by MAP Bayesian fitting.
`The plasma PA and PG disposition curves were also ana-
`
`Downloaded from
`
`on August 3, 2018. © 2001 American Association for Cancerclincancerres.aacrjournals.org
`
`
`Research.
`
`Par Pharmaceutical, Inc. Ex. 1007
`Par v. Horizon, IPR of Patent No. 9,561,197
`Page 4 of 10
`
`

`

`Table 3 No. of patients with grade 3 or 4 toxicity by dose level
`
`150
`mg/kg/d
`
`225
`mg/kg/d
`
`285
`mg/kg/d
`
`410
`mg/kg/d
`
`345
`mg/kg/d
`1
`
`1b
`
`2c
`
`Neurocortical
`Hypokalemia
`Hyperuricemia
`Nausea
`Neutropenia
`Fatigue
`a Toxicities occurred in the same patient.
`b Lasted 2 days only.
`c Felt to be related to disease progression rather than drug.
`
`515
`mg/kg/d
`1a
`1a
`1a
`1a
`
`Clinical Cancer Research
`
`3051
`
`emetics, such as the phenothiazines (prochloperazine). This
`observation led to the use of nonsedating antiemetics such as the
`serotonin 5-HT3 receptor antagonists, ondansetron and granis-
`etron. Patients had less sedation and minimal emesis with this
`change in antiemetic.
`Hematological Toxicity. No dose-limiting hematologi-
`cal toxicity was noted. Minor decreases in hematocrit were
`noted over the course of therapy. No platelet toxicities were
`noted in any patient. One patient had a transient grade 3 neu-
`tropenia (absolute neutrophil count of 600) noted at dose level 1
`that resolved in 2 days. No other declines in total white cell
`count or in subpopulations were noted.
`Nonhematological Toxicity. Patients experienced mild
`grade 2 nausea at the higher dose levels, in particular, the 410
`mg/kg/day dose. Fatigue was dose related as well. Grade 2
`fatigue was noted in 1 patient at 345 mg/kg/day and 2 patients
`at 410 mg/kg/day. Grade 3 fatigue was noted in 2 of 7 patients
`receiving 410 mg/kg/day infusions. This degree of fatigue was
`not felt to be drug related but rather attributable to tumor
`progression. In fact, the 2 patients with grade 3 fatigue did not
`receive a rechallenge with PB as they were taken off study for
`progressive disease. Of note, 7 (38%) of the patients with bony
`metastatic PCA experienced a flare in bone pain that started on
`discontinuation of the infusion and persisted for 48 h. Only the
`2 patients with grade 3 neuro-cortical toxicity developed the
`characteristic odor of PA. The ability to detect the odor also
`resolved promptly with discontinuation of the study drug.
`Pharmacokinetics and Pharmacodynamics. Of 24 pa-
`tients enrolled in the study, 1 patient had incomplete blood
`sampling for pharmacokinetic data analysis, and 2 patients did
`not have adequate plasma sample volumes to allow determina-
`tion of PA and PG concentrations. These 3 patients were ex-
`cluded from the pharmacokinetic data analysis.
`In the majority of the patients, the plasma PB disposition
`curves were characterized by a rise to an apparent plateau
`achieved by 4 – 6 h into the infusion. However, a number of the
`subjects had curves that showed a substantial decline in PB
`concentrations starting around 24 h into the infusion and con-
`tinuing throughout the remainder of the infusion. This pattern
`was seen in 0 of 4 patients 150 mg/kg/day, 0 of 4 patients at 225
`mg/kg/day, 1 of 4 patients at 285 mg/kg/day, 3 of 4 patients at
`345 mg/kg/day, 5 of 7 patients at 410 mg/kg/day, and 0 of 1
`patient at 515 mg/kg/day. This pattern suggested that the plasma
`clearance of PB increased in a continuous fashion beginning
`24 h into the infusion. Fig. 2 demonstrates the PB disposition
`pattern for 2 patients, 1 at 225 mg/kg/day and the other at 515
`mg/kg/day. This feature was incorporated in the pharmacoki-
`netic models used to describe the disposition curves.
`Individual subject PB disposition curve analysis using a
`pharmacokinetic model with first-order elimination showed an
`inverse relationship between clearance rate and dose, suggesting
`that a model with saturable elimination would be more appro-
`priate. In the model that was used in the population pharmaco-
`kinetic analysis of PB disposition, elimination was modeled as
`being Michaelis-Menten in character. The population pharma-
`cokinetic estimates for PB disposition are listed in Table 4. The
`mean kinc value of 0.0029/h equates to a 1.32-fold increase in
`Vmax by 120 h into the infusion.
`The population pharmacokinetic estimates for PA and PG
`
`One patient refused additional therapy after six cycles for per-
`sonal reasons despite continued stable disease. Another patient
`discontinued therapy secondary to toxicity during his first cycle
`of therapy at 410 mg/kg/day. Lastly, 1 patient had toxicity at
`two different dose levels despite dose reduction and met criteria
`for withdrawal secondary to toxicity. This patient had a 10%
`decline in his PSA and subjective improvement in bone pain
`over the two cycles that he received therapy.
`Three patients had their dose escalated during the course of
`therapy. Two of these patients remained on the study for a total
`of eight cycles. In fact, 1 of these patients had his dose increased
`twice from 150 mg/kg/day (dose level 1) to 285 mg/kg/day
`(dose level 3). Two patients had their dose reduced secondary to
`toxicity at their starting dose during cycle 1. The starting doses
`for these patients were 345 and 515 mg/kg/day. No patient at
`410 mg/kg/day had their dose reduced or escalated and was
`determined to be the recommended Phase II dose. Table 3
`describes all grades 3 and 4 toxicities during the study period.
`Neuro-cortical Toxicity. DLT was neuro-cortical in na-
`ture. One patient on 515 mg/kg/day experienced grade 3 neuro-
`cortical toxicity ⱕ48 h of the start of drug infusion. The toxicity
`was exemplified by excessive somnolence and confusion and
`was accompanied by metabolic changes consisting of grade 4
`hypokalemia (2.1 mmol/liter), grade 3 hyponatremia (128
`mmol/liter), grade 2 hypocalcemia (7.9 mg/dl), and grade 4
`hyperuricemia (15.2 mg/dl). The metabolic nadirs occurred 4
`days after initiation of infusion. The patient also had grade 3
`nausea before the decline in mental status. Full recovery and
`return to baseline clinical state occurred ⱕ10 –12 h after dis-
`continuation of the drug at approximately the same time as the
`electrolyte abnormalities returned to normal values after aggres-
`sive repletion. The patient received a second cycle at the next
`lower dose and experienced a similar toxicity syndrome on
`rechallenge. An additional patient developed grade 3 neuro-
`cortical toxicity at the 345 mg/kg/day dose. This was not ac-
`companied by electrolyte abnormalities and occurred at 96 h
`into the infusion. The symptoms resolved 10 –12 h after discon-
`tinuing the drug. A 3rd patient developed grade 2 neuro-cortical
`toxicity with only grade 1 hyponatremia and hypocalcemia at
`410 mg/kg/day. This patient was not rechallenged at a lower
`dose because he developed obstructive urinary symptoms and
`was taken off study for progression of disease. Of note, plasma
`ammonia levels in the grade 3 toxicity patients remained in the
`normal range.
`Patients with nausea had more sedating effects from anti-
`
`Downloaded from
`
`on August 3, 2018. © 2001 American Association for Cancerclincancerres.aacrjournals.org
`
`
`Research.
`
`Par Pharmaceutical, Inc. Ex. 1007
`Par v. Horizon, IPR of Patent No. 9,561,197
`Page 5 of 10
`
`

`

`3052 Phase I Study of Phenylbutyrate
`
`Table 4 Population pharmacokinetic estimatesa
`
`Mean
`
`SD
`
`0.169
`148
`0.0029
`546
`
`PB (n ⫽ 20)
`volume of distribution, liter/kg
`Vmax, ␮mol/h/kg
`kinc, 1/h
`km, ␮mol/liter
`PA (n ⫽ 19)
`volume of distribution, liter/kg
`Vmax, ␮mol/h/kg
`kinc, 1/h
`km, ␮mol/liter
`Phenylacetylglutamine (n ⫽ 19)
`0.281
`volume of distribution, liter/kg
`1.83
`clearance rate, ml/min/kg
`a Kinc, rate constant of increase in Vmax after 24 h.
`
`0.720
`77
`0.0009
`62
`
`0.043
`23
`0.0015
`159
`
`0.562
`10
`0.0006
`28
`
`0.130
`1.15
`
`collections in which the calculated urine clearance rates were
`unphysiologically large, the mean urine clearance rate for the
`0 –24-h collection period is 116 ml/min (SD, 62 ml/min), and for
`the 96 –120-h collection period, it is 136 ml/min (SD, 97 ml/
`min). The coefficient of correlation for paired values is 0.84. A
`paired t test indicated that the increase in the mean value of the
`urine clearance rate between the first and second urine collec-
`tions is not significant (P ⫽ 0.10, two-tailed test). Previous
`studies have shown that PB is almost completely metabolized to
`PA via ␤-oxidation

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