throbber
CENTER FOR DRUG EVALUATION AND
`
`CENTER FOR DRUG EVALUATION AND
`RESEARCH
`RESEARCH
`
`
`
`
`
`
`APPLICATION NUMBER:
`22-272
`22-272
`
`
`
`APPLICA TION NUMBER:
`
`CLINICAL PHARMACOLOGY AND
`
`CLINICAL PHARMACOLOGY AND
`BIOPHARMACEUTICS REVIEW(S)
`BIOPHARMACEUTICS REVIEW! S 2
`
`
`
`
`
`

`

` PRE-IND
`ANIMAL to HUMAN SCALING
` IN-VITRO METABOLISM
` PROTOCOL
` PHASE II PROTOCOL
` PHASE III PROTOCOL
` DOSING REGIMEN CONSULT
` PK/PD- POPPK ISSUES
` PHASE IV RELATED
`
`
`
`
`
`
`
`
`
` NAI (No action indicated)
` E-mail comments to:
`Medical Chemist
`Pharm-Tox
`Micro
`Pharmacometrics Others
`(Check as appropriate and attach e-mail)
`
`
` DISSOLUTION/IN-VITRO RELEASE
` BIOAVAILABILITY STUDIES
` IN-VIVO WAIVER REQUEST
` SUPAC RELATED
` CMC RELATED
` PROGRESS REPORT
` SCIENTIFIC INVESTIGATIONS
` MEETING PACKAGE (EOP2/Pre-
`NDA/CMC/Pharmacometrics/Others)
`
` FINAL PRINTED LABELING
` LABELING REVISION
` CORRESPONDENCE
` DRUG ADVERTISING
` ADVERSE REACTION REPORT
` ANNUAL REPORTS
` FAX SUBMISSION
` OTHER (SPECIFY BELOW):
`DDI study protocol employing Oxycontin 10
`mg (ER) with 200 mg bid ketoconazole
`
`
`
`REVIEW ACTION
` Oral communication with
`Name: [ ]
` Comments communicated in
`meeting/Telecon. see meeting minutes
`dated: [ ]
`
`
`
` Formal Review/Memo (attached)
`See comments below
`See submission cover letter
` OTHER (SPECIFY BELOW):
` [ ]
`
`
`
`DEPARTMENT OF HEALTH AND
`HUMAN SERVICES
`PUBLIC HEALTH SERVICE
`FOOD AND DRUG ADMINISTRATION
`
`
`From: Sheetal Agarwal, Ph.D.
`
`
`DATE: 04/29/09
`
`
`IND No.: 29,038
`SDN.:687
`
`
`NAME OF DRUG: Oxycontin
`
`NAME OF THE SPONSOR: Purdue Pharma
`
`
`
`Related NDA Nos.
`20-553 (SDN 351),
`22-272 (SDN 60)
`
`PRIORITY CONSIDERATION
`
`Clinical Pharmacology
`Tracking/Action Sheet for Formal/Informal Consults
`
`To: DOCUMENT ROOM (LOG-IN and LOG-OUT)
`Please log-in this consult and review action for the specified
`IND/NDA submission
`
`Submission Date: 04/16/2010
`
`
`
`
`
`Date of informal/Formal Consult:
`
`
`
`
`TYPE OF SUBMISSION
`
`CLINICAL PHARMACOLOGY/BIOPHARMACEUTICS RELATED ISSUE
`
`
`
`REVIEW COMMENT(S)
` NEED TO BE COMMUNICATED TO THE SPONSOR
` HAVE BEEN COMMUNICATED TO THE SPONSOR
`
`
`
`
`COMMENTS/SPECIAL INSTRUCTIONS:
`
`The submitted study protocol OTR1023 for a drug-drug interaction study employing Oxycontin and
`ketoconazole as a Phase 4 post marketing commitment for approval of Oxycontin is acceptable from a
`Clinical Pharmacology perspective. No further action is indicated at this time.
`
`BACKGROUND:
`
`This review pertains to the final drug-drug interaction protocol # OTR1023 submitted to the Agency on
`04/16/2010. This DDI study is designed to fulfill the Post Marketing Commitment outlined in the Agency’s
`letter dated September 2, 2009 in reference to the approval of supplement # S-060 that was submitted
`on December 13, 2007. A draft of protocol OTR1023 was submitted on September 11, 2009 (NDA
`20533/SDN 340) and reviewed by Dr. Sayed Al Habet (see review in DARRTS dated 09/30/00).
`
`
`
`
`

`

`
`
`The following Clinical Pharmacology related comment was conveyed to the sponsor at the time of initial
`review of the draft protocol:
`
`“Although, a drug interaction is expected between ketoconazole and oxycontin, the magnitude of
`resulting increase in oxycodone exposure is unknown. In order to protect the healthy volunteers
`participating in this study from the opioid side effects resulting from a potential interaction, we advice that
`you provide naltrexone blockade to the participating volunteers. We recommend that naltrexone at a
`dose of 50 mg be administered during the study at the following time points in relation to oxycontin
`dosing: 12 hours pre-dose, 12 hours, 24 and 36 hours post-dose.”
`
`It should be noted that the final protocol does not contain any major amendments to the study design
`reviewed by Dr. Al Habet which would materially affect the clinical pharmacology assessments.
`
`
`Purdue did not include naltrexone blockade in their final protocol but provided an acceptable rationale to
`the Agency via email on April 27, 2010 to the project manager, Ms. Lisa Basham. Dosing in this study
`was planned to be started on April 28, 2010. Attachment 1 is an extract of Purdue’s rationale, while
`attachment 2 contains the final protocol synopsis.
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`

`

`
`ATTACHMENT 1: RATIONALE PROVIDED BY PURDUE PHARMA FOR NOT INCLUDING
`ATTACHMENT 1: RATIONALE PROVIDED BY PURDUE PHARMA FOR NOT INCLUDING
`NALTREXONE BLOCKADE IN THE DRUG-DRUG INTERACTION STUDY
`NALTREXONE BLOCKADE IN THE DRUG-DRUG INTERACTION STUDY
`
`
`
`jApril26,2fl1flj FDA Comment on proposed studv OTR1U23:
`
`“Aithough, a drug interaction is expected between hetoconazoie and vaflontin, the magnitude of
`
`resulting increase in oxvcodone exposure is unknown. in order to protect the healthy voiunteers
`
`participating in this studvfrom the opioid side effects resuiting Jfrom a potentiai interaction, we advise
`
`that you provide naltrexone blockade to the participating volunteers. We recommend that naitrexone at
`
`a dose of Eli] ma be administered during the study at the Jfoiiowingr time points in reiation to OxvContin
`
`dosing: 1.2 hours pre—dose, 12 hours. 2:11 anaI 35 hours post—dose.”
`
`April 2? 2pm PPLP Response:
`
`OTR1023 is a drug—drug interaction studv in which 10 mg OxvContin (oxvcodone hvdrochloride
`
`controlled—release [CR]]I tablets will be administered to subjects with and without concomitant
`
`ketoconazole administration to assess the impact of this potent azole CYP3A4 inhibitor on oxvcodone
`
`pharmacokinetics. We considered inclusion of naltrexone blockade in protocol DTR1D23 but elected not
`
`to include it based upon the considerations summarized below.
`
`In a published report, Hagelberg et al (Euri Clin Pharmacol. 20¢]?! Mar:65{3}:263-?1 attached in email]
`
`examined the interaction between oxvcodone and the potent azole CYPSAA inhibitor voriconazole in 12
`
`healthv subjects. vacodone was administered as single 10 mg immediate-release {le capsule
`
`{Oxvnormj doses. In the presence of voriconazole. mean peak oxvcodone (Cmaxj increased from 18.1 to
`
`3&5 ngimL. This corresponds to a 12-fold increase in Cmax on average {range 1.4 —2.4x). Mean total
`
`oxvcodone exposure {AUCinfj increased from 1:32 to 363 ng.himL. This corresponds to a 16—fold
`
`increase in AUCinf on average [range 2.? — 5.6x].
`
`Adverse events were described bv Hagelberg et al as follows:
`
`r’AII subjects completed the studv. Eight of the 12 subjects experienced adverse
`
`events on dav 3. Adverse events were headache (n=5]. nausea {n=3]I, vomiting
`
`{n21}. dizziness {n22}. extremefatigue [n21] and itch {n21}. Three subjects
`
`received paracetamol [1,0130 mg} for headache 12 h after oxvcodone dosing. and
`
`one received tropisetrone 2 mg iv for nausea 5 h after dosing. All cases of nausea
`
`or vomiting were reported during the voriconazole phase. Number of reports of
`
`headache did not differ between voriconazole and control phases.”
`
`PPLP concluded that although the increase in oxvcodone exposure following co-administration with
`
`voriconazole was associated with more AEs, there were no significant safetv concerns raised bv the
`
`observed AEs following the administration of 10 mg IR oxvcodone under CYP3A4 inhibition, bevond
`
`those applicable whenever an opioid is administered under experimental conditions.
`
`We hvpothesize that since ketoconazole and voriconazole are both potent azole CYP3A4 inhibitors. thev
`
`will have similar effects on oxvcodone pharmacokinetics. We further hvpothesize that the magnitudes of
`
`the increases in Cmax and AUC. noted bv Hagelberg et al are the best available predictions of the
`
`magnitude of effect likelv to be characterized in OTR1023. These hvpotheses are supported bv observed
`
`
`
`
`
`

`

`
`
`effects of these C‘r’P3fit4 inhibitors on sirolimus exposure in healthy subjects. ‘y'oriconazole produced
`
`increases in sirolimus exposure of 2—fold and 11—fold for Cmax and AUC, respectiyely jyfend Package
`
`Insert attaached in email]. Ketoconazole produced increases in sirolimus exposure of 4.4—fold and 11—
`
`fold for Cmax and fiUC, respectiyely [Floren et al. Clin Pharm Ther [1999] 65, 159—159 attached in
`
`email].
`
`In OTR1923, oxycodone is administered in CR form as a 19 mg nyContin dose. In the absence of
`
`CYP3A4 inhibition, this dose is expected to produce a mean Cmax of approximately 9.4 ngme and an
`
`EUCinf of approximately 198 ng.h;'mL. It should be noted that this expected peak exposure [Cmax] is
`
`approximately 59% of that expected for a 19 mg IR oxycodone dose, while total oxycodone exposure
`
`{fiUCinfl is similar for ER and IR formulations. Thus, use of a CR dosage form [OxyContin] proyides a 2—
`
`fold reduction in expected Cmax, with and without CYP3A4 inhibition. Since the intensity of opioid AEs is
`
`typically related to lEmax, this margin is releyant to the safety and tolerability of oxycodone closing in
`9T91923.
`
`In a prior PPLP single—dose crossoyer study DC93—9891 [submitted to IND 29,938 on October 2, 1994,
`
`Serial Number 188]], both 29 and 49 mg nyContin doses and 29 mg IR oxycodone doses were
`
`administered to healthy subjects {n=24] without naltrexone blockade. Mean oxycodone Cmax and EU:
`
`following 49 mg nyContin administration were 39.3 ngme {range 23.9 — 92.5] and 421 ng.h;'mL {range
`
`244 — 921], respectiyely. The l9993—9891 study report states that most .4Es were mild or moderate in
`
`intensity and that there were no discontinuations due to adyerse experiences. It further states that a
`
`dose—response was obseryed between 29 mg {n=22] and 49 mg {n=24] lEixyilontin doses, with 9? and
`
`19? AEs reported, respectiyely, for the two treatments.
`
`The prior safety and tolerability experience in l9993—9891 with 49 mg nyContin administered without
`
`naltrexone represents the safety and tolerability that is expected in l9TH1923 assuming ketoconazole
`
`were to produce approximately a 4-fold increase in Cmax fits. the 1.?-fold increase noted with
`
`yoriconazolej and AUC jys. the 3.6—fold increase noted with yoriconatole].
`
`fidministration of 59 mg naltrexone blockade is belieyed by our inyestigators to be associated with
`
`tolerability issues, reflected by reported AEs, and can eyen lead to subject discontinuations in rare
`
`instances.
`
`While exclusion of naltrexone is adyantageous in permitting a “cleaner” assessment of the effect of
`
`ketoconazole on oxycodone pharmacokinetics, this consideration only applies if the conclusion is
`
`reached that naltrexone blockade is not required to minimise the opioid agonist effects anticipated in
`
`this study. Based upon the considerations summarised aboye, we concluded that naltrexone blockade is
`
`not required in this study. Therefore co—administration of naltrexone was not included in protocol
`
`[2991923.
`
`
`
`
`
`.
`
`
`
`
`
`

`

`
`ATTACHMENT 2: FINAL PROTOCOL SYNOPSIS
`
`
`
`
`
`
`
`
`(b) (4)
`
`

`

`Treatments, Doses, and Modes of Administration:
`
`OTR 10 mg tablet, Ketoconazole 288 mg oral tablet, and placebo tablet {administered o12h]. All treatments
`
`will be administered orally with 8 oz. {248 mL] water.
`
`Treatments will be administered in an open-label fashion. Subjects will be dosed in an upright position, and
`
`will remain upright for4 hours afterwards. Treatment administrations will be separated by a 14 day
`
`washout period.
`
`Concomitant Medication:
`
`placebo or upon discontinuing from the study.
`
`Naloxone HCI challenge test (administered on Day —1, Period 1 check-in].
`
`The use of concomitant medications during this trial is discouraged, unless necessary to treat adverse
`events or unless approved on a case—by—case basis prior to randomization leg, hormonal contraceptives].
`The use of any concomitant medications should be approved by the sponsor in advance, in writing, when
`possible.
`
`Duration of Treatment and Study Duration:
`
`Screening and baseline period will be up to 28 days prior to administration of study medication (in this case
`either ketoconazole or placebo administration).
`
`In Period 1 subjects will be administered ketoconazole or placebo in a randomized fashion on days 1-4.
`On day 3, the subjects will receive a single oral dose of OTR. This will be followed by a washout period
`lasting 14 days {days 5 to 18}. In Period 2 subjects will be administered ketoconazole or placebo in a
`crossover fashion on days 18—22. On day 21, the subjects will receive a single oral dose of OTR.
`
`Subjects will be confined to the study facility during Periods 1 and 2.
`
`Subjects will have end of study procedures fEOS} performed T—1 8 days after last dose of ketoconazole or
`
`The total duration ofthe study is approximately 88 days.
`
`Treatment Schedule:
`
`Pro-Randomization Phase:
`
`Screening: Subjects will be screened within 28 days of Period 1 check—in. Drug and alcohol screens,
`physical exam, 12— lead ECO, vital signs (systolic blood pressure, diastolic blood pressure, pulse rate,
`respiration rate and oral temperature), SpOg, medical and medication history, clinical laboratory testing and
`inclusion-“exclusion criteria will be evaluated. Subjects are not allowed to consume applejuice, orange juice
`or grapefruit juice during the treatment period.
`
`Randomization Phase:
`
`On Day -1, Period 1 Check-in only, subjects will receive a Naloxone HOI challenge test. Vital signs and
`SpOg will be measured prior to and following the Naloxone HCI challenge test.
`
`For each Period, subjects will check into the unit the day prior to dosing. At check—In, subjects will have
`chemistry (fasting for at least 4 hours}, hematology and urinalysis tests performed. Urine pregnancy test
`{for women of childbearing potential], vital signs, SpOE, and alcohol and urine drug screens will be
`performed.
`
`Period 1 (Days 1 to 5}
`
`Ketoconazole {288 mg} or placebo administration will begin on Day 1 and will be given twice a day at
`approximately 8 Alt-1 and approximately 8 Plvl through Day 4. Throughout Period 1, vital signs, SpOg and
`HDYF? will be performed as per study flow chart.
`
`On Day 3 at approximately 8 AM, OTR 18 mg tablet will be administered following an overnight fast.
`Subjects will continue fasting until 4 hours post—dose. Blood samples for PK analysis will be drawn at pre-
`dose, and at ore-determined time points through 48 hours post—dose.
`
`
`
`
`
`
`

`

`On Day Ll: blood samples for the determination of ketoconazole levels will be drawn at approximately 8 Alvl,
`prior to ketoconazole or placebo administration.
`
`On Day 5: vital signs, SpOg and HDYF? will be performed as per study flow chart and the subject will then
`be discharged with instructions to return on Day ‘I S.
`
`Wash-out Period {Days 5 to 13}
`
`Baseline 2 [Day 181
`
`During this period, the investigator will ensure that provisions are made for the subjects to contact the study
`site in case of adverse events during the washout period. Such reporting ofadverse events and the
`investigators response will be accurately documented, and the Purdue Pharma L.P. study monitor will be
`notified immediately.
`
`analysis set will be documented in the Statistical Analysis Plan.
`
`‘v’ital sign evaluation, safety laboratory tests {serum
`On Day 13, subjects will be confined to the study unit.
`chemistries including liver function tests, hematology, and urinalysis}; urine pregnancy testing (if female],
`urine drug screening: and alcohol testing will be performed.
`
`Period 2 (Days 19 to 23)
`
`On Day '19, the subjects will be crossed over to the opposite treatment schedule. Subjects who were given
`ketoconazole during Period 1 will be given placebo in Period 2, and those given placebo in Period "i will be
`given ketoconazole during Period 2.
`
`Ketoconazole {200 mg) or placebo administration will begin on Day 19 and will be given twice a day at
`approximately 8 AM and approximately 8 PM through Day 22. Throughout Period 2, vital signs, SpOg and
`HDYF? will be performed as per study flow chart.
`
`On Day 21 at approximately 8 Alvl, OTR 113 mg tablet will be administered following an overnight fast.
`Subjects will continue fasting until 4 hours post—dose. Blood samples for PK analysis will be drawn at pre-
`dose: and at ore-determined time points through 48 hours post—dose.
`
`On Day 22, blood samples for the determination of ketoconazole levels will be drawn at approximately 8
`AM, prior to OTR and ketoconazole administration.
`
`On Day 23. vital signs: SpO2 and HDYF‘? will be performed as per study flow chart and the subject will then
`be discharged with instructions to return for End of Study procedures.
`
`Adverse events {AEs} and concomitant medications will be recorded throughout the study.
`
`Note: for clarity: OTR is assigned as the study drug for determination of adverse event causality.
`Ketoconazole and placebo are considered distinct from study drug in this determination.
`
`End of Study Visit: Subjects will return to the unit T to it} days after receiving their last dose of
`ketoconazole or placebo or upon discontinuation from the study for their end of study {EOS} procedures.
`EOS procedures will include a physical exam, ’12- lead ECG. vital signs, SpOg: and laboratory tests.
`
`Criteria and Methods for Evaluation:
`
`Analysis Pogulations:
`
`The enrolled gogulation is the group of subjects who provide informed consent.
`
`The randomized safety gogulation is the group of subjects who are randomized, receive study drug. and
`have at least one post dose safety assessment.
`
`The full analysis gopulation for PK metrics will be the group of subjects who are randomized, receive study
`drug, and have at least one valid PK metric for that treatment. Subjects experiencing emesis within 12
`hours after dosing may be excluded from PK analysis. Subjects and profiles-"metrics excluded from the
`
`
`
`
`
`
`
`

`

`Oxycodone Concentration Measurements: Ellood samples for determining the concentrations of oxycodone
`and its metabolites {noroxycodone oxymorphone, and noroxymorphonej in plasma, will be obtained for
`each subject at predose and at 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 10, 12, 16,24, 28, 32, 38, and 48 hours
`post study drug administration during each ofthe study Periods.
`
`Elioanalytical methods:
`
`Plasma oxycodone and its metabolites (noroxycodone, oxymorphone, and noroxymorphoneji
`concentrations will be quantified using a validated liquid chromatography tandem mass spectrometric
`method.
`
`Safety Assessments: Safety will be assessed using recorded adverse events, clinical laboratory test
`results, vital signs, SpCrg, physical examinations, and electrocardiograms (ECG).
`
`Statistical Methods:
`
`
`
`Su bjects‘ AEs will be categorized into preferred terms and associated system organ class using the
`Medical Dictionary for Regulatory Activities (h-ledDRA}. Treatment—emergent AEs will be defined as AEs
`that start after, or increase in severity after, a dose of study drug. Treatment—emergent AEs that occur
`during the washout period up to the point of dosing the next study drug will be assigned to the previous
`treatment dosed. Treatment—emergent AEs will be summarized by presenting the incidence ofAEs for each
`treatment group by the MedDRA preferred term, nested within System Organ Class for the safety
`population.
`
`Pharmacokinetic Metrics: Plasma concentrations of oxycodone and its metabolites (noroxycodone,
`oxymorphone, and noroxymorphone} will be analyzed to determine the following pharmacokinetic metrics:
`AUCt, AUCinf, Cmax, tmax, tlag, t‘|.-’22, and LambdaZ. Descriptive statistics will be tabulated by treatment,
`as applicable, for all pharmacoltinetic metrics.
`
`Safety Analysis:
`
`All safety data (AEs, clinical laboratory results, vital signs, SpClg and ECGs) will be listed for subjects in the
`enrolled and safety populations. Results of clinical laboratory evaluations that lie outside the normal range
`will be flagged on the listings as high or low.
`
`Laboratory, vital signs and 8pc}; will be summarized by treatment and time—point for the safety population.
`
`Sample Size Rationale:
`
`No formal sample-size calculations were performed. A sufficient number of subjects {up to 3D) will be
`randomized to complete approximately 20 subjects.
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`

`

`Application
`Type/Number
`--------------------
`NDA-22272
`
`Submission
`Type/Number
`--------------------
`PMR/PMC-1
`
`Submitter Name
`
`Product Name
`
`--------------------
`PURDUE PHARMA
`INC
`
`------------------------------------------
`OXYCONTIN
`
`---------------------------------------------------------------------------------------------------------
`This is a representation of an electronic record that was signed
`electronically and this page is the manifestation of the electronic
`signature.
`---------------------------------------------------------------------------------------------------------
`/s/
`----------------------------------------------------
`
`SHEETAL S AGARWAL
`04/30/2010
`
`SURESH DODDAPANENI
`04/30/2010
`
`

`

` PRE-IND
`ANIMAL to HUMAN SCALING
` IN-VITRO METABOLISM
` PROTOCOL
` PHASE II PROTOCOL
` PHASE III PROTOCOL
` DOSING REGIMEN CONSULT
` PK/PD- POPPK ISSUES
` PHASE IV RELATED
`
`
`
`
`
`
`
`
`
` NAI (No action indicated)
` E-mail comments to:
`Medical Chemist Pharm-Tox
` Micro Pharmacometrics Others
`(Check as appropriate and attach e-
`mail)
`
`
` DISSOLUTION/IN-VITRO
`RELEASE
` BIOAVAILABILITY STUDIES
` IN-VIVO WAIVER REQUEST
` SUPAC RELATED
` CMC RELATED
` PROGRESS REPORT
` SCIENTIFIC INVESTIGATIONS
` MEETING PACKAGE (EOP2/Pre-
`NDA/CMC/Pharmacometrics/Others)
`REVIEW ACTION
` Oral communication with
`Name: [ ]
` Comments communicated in
`meeting/Telecon. see meeting minutes
`dated: [ ]
`
`
`
` FINAL PRINTED LABELING
` LABELING REVISION
` CORRESPONDENCE
` DRUG ADVERTISING
` ADVERSE REACTION REPORT
` ANNUAL REPORTS
` FAX SUBMISSION
` OTHER (SPECIFY BELOW):
`Labeling changes related to drug-drug
`interactions following published reports
`
`
`
` Formal Review/Memo (attached)
`See comments below
`See submission cover letter
` OTHER (SPECIFY BELOW):
` [ ]
`
`
`
`DEPARTMENT OF HEALTH AND
`HUMAN SERVICES
`PUBLIC HEALTH SERVICE
`FOOD AND DRUG ADMINISTRATION
`
`
`From: Sheetal Agarwal, Ph.D.
`
`
`IND No.:
`Serial No.:
`
`
`DATE: 03/15/10
`
`
`NAME OF DRUG
`Oxycontin®
`
`NAME OF THE SPONSOR: Purdue Pharma
`
`
`Clinical Pharmacology
`Tracking/Action Sheet for Formal/Informal Consults
`
`To: DOCUMENT ROOM (LOG-IN and LOG-OUT)
`Please log-in this consult and review action for the
`specified IND/NDA submission
`
`Submission Date: 02/05/10
`
`
`
`
`
`PRIORITY
`CONSIDERATION
`
`
`
`Date of informal/Formal
`Consult:
`
`
`
`
`
`NDA No. 22-272
`SDN: 50
`
`TYPE OF SUBMISSION
`CLINICAL PHARMACOLOGY/BIOPHARMACEUTICS RELATED ISSUE
`
`
`
`REVIEW COMMENT(S)
` NEED TO BE COMMUNICATED TO THE SPONSOR
` HAVE BEEN COMMUNICATED TO THE SPONSOR
`BACKGROUND:
`
`This is a Class I resubmission of NDA 22272 pertaining to reformulated oxycontin formulation. The original
`NDA submission and the complete response submission were previously reviewed by Dr. Sayed Al Habet
`(see reviews dated 5/23/2008 and 9/1/2009 for complete details regarding clinical pharmacology aspects of
`this product). Pertinent Clinical Pharmacology relevant labeling language was negotiated with the sponsor in
`the last previous cycle. However, subsequent to this, Drug-Drug interaction data between coadministration of
`oxycodone and rifampin from the following publication came to light; Nieminen TH et al. Rifampin greatly
`reduces the plasma concentrations of intravenous and oral oxycodone. Anesthesiology.
`2009;110:1371-1378.
`
`This review captures the labeling changes related to this interaction and other minor appropriate
`modifications suggested by this reviewer. Attachment contains the proposed labeling changes (additions
`indicated by underlined text and deletions indicated by strikethrough text).
`
`Following is a discussion of the rifampin and oxycodone interaction study. In this four-session, paired,
`crossover study, twelve volunteers were given 600 mg oral rifampin or placebo once daily for 7 days.
`
`

`

`Oxycodone was given on day 6. In the first part of the study, 0.1 mg/kg oxycodone hydrochloride was given
`intravenously. In the second part of the study, 15 mg oxycodone hydrochloride was given orally.
`Concentrations of oxycodone and its metabolites noroxycodone, oxymorphone, and noroxymorphone were
`determined for 48 h. Psychomotor effects were characterized for 12 h by several visual analog scales.
`Analgesic effects were characterized by measuring the heat pain threshold and cold pain sensitivity.
`
`Plasma profiles and PK parameters are listed in Figure 1 and Tables 1 and 2 extracted from the publication.
`
`Rifampin decreased the AUC of intravenous oxycodone by 53%, increased the mean plasma Cl by 2.2-fold,
`and decreased its t½ from 3.7 to 2.4 h. Rifampin decreased the mean AUC and Cmax of oral oxycodone by
`86% and 63%, respectively. The mean oral bioavailability of oxycodone was decreased from 69% to 21% by
`rifampin. Rifampin increased the Cmax of noroxycodone by 2.7-fold after intravenous oxycodone and by 2.0-
`fold after oral oxycodone compared with the control values. The corresponding metabolite–to–parent drug
`AUC ratios (AUCm/AUCp) were increased 2.4-fold and 7.6-fold, respectively. Rifampin reduced the AUC of
`oxymorphone to approximately 5–10% of the corresponding control value after intravenous and oral
`administration of oxycodone. Intravenous oxycodone produced no detectable oxymorphone concentrations in
`two subjects after placebo and in eight subjects after rifampin. After rifampin and oral oxycodone, five of the
`subjects had every measured oxymorphone concentration below the lower limit of quantification. Rifampin
`increased the Cmax of noroxymorphone more than twofold in both the intravenous and the oral part of the
`study. The corresponding metabolite– to–parent drug AUC ratios increased 2.4-fold after intravenous and
`9.6-fold after oral oxycodone. In addition, pharmacologic effects of oral oxycodone were attenuated.
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`

`

`Figure 1: Plasma concentrations (mean - SD) of oxycodone and its metabolites in 12 volunteers following
`intravenous administration of 0.1 mg/kg and oral administration of 15 mg oxycodone hydrochloride
`after placebo or rifampin. The volunteers were given in randomized order either 600 mg oral rifampin or
`placebo once daily at 8 PM for 7 days. Oxycodone was given on day 6 at 8 AM, 12 h after the fifth dose
`of rifampin or placebo. Two more doses of rifampin or placebo were given on days 6 and 7. The
`oxycodone concentrations are shown both on arithmetic and on a semilogarithmic plot (inset).
`
`
`
`
`
`
`
`
`
`
`
`

`

`Table 1: Pharmacokinetic Parameters of Oxycodone and Its Metabolites in Volunteers after Intravenous
`Administration of Oxycodone Following Placebo (Control) or Oral Rifampin
`
`
`
`Table 2: Pharmacokinetic Parameters of Oxycodone and Its Metabolites in Volunteers after Oral
`Administration of Oxycodone Following Placebo (Control) or Oral Rifampin
`
`
`
`
`
`
`
`
`

`

`5 WARNINGS AND PRECAUTIONS
`
`5.8 Cytochrome P450 3A4 Inhibitors and Inducers
`
`Since the CYP3A4 isoenzyme plays a major role in the metabolism of OxyContin, drugs
`that alter
` CYP3A4 activity may cause changes in
` clearance of
`oxycodone which could lead to an increase or decrease in oxycodone plasma
`concentrations. The expected clinical results with CYP450 inhibitors would be increased
`or prolonged opioid effects. If co-administration with OxyContin is necessary, caution is
`advised when initiating therapy in patients, currently taking, or discontinuing CYP3A4
`inhibitors. Evaluate these patients at frequent intervals and consider dose adjustments
`until stable drug effects are achieved. The expected clinical results with CYP450 inducers
`would be decrease in oxycodone plasma concentrations, lack of efficacy or, possibly,
`development of abstinence syndrome in a patient who had developed physical
`dependence to oxycodone. Evaluate these patients at frequent intervals and consider dose
`adjustments until stable drug effects are achieved. [see Drug Interactions (7.2) and
`Clinical Pharmacology (12)]
`
`
`7.1 Neuromuscular Junction Blocking Agents
`
`OxyContin may enhance the neuromuscular blocking action of true skeletal muscle
`relaxants (such as pancuronium) and produce an increased degree and/or duration of
`respiratory depression.
`
`7.2 Agents Affecting Cytochrome P450 Isoenzymes
`
`Inhibitors of CYP3A4:
`Since the CYP3A4 isoenzyme plays a major role in the metabolism of OxyContin, drugs
`that inhibit CYP3A4 activity, such as macrolide antibiotics (e.g., erythromycin), azole-
`antifungal agents (e.g., ketoconazole), and protease inhibitors (e.g., ritonavir), may cause
`decreased clearance of oxycodone which could lead to an increase in oxycodone plasma
`concentrations. A published study showed that the co-administration of the antifungal
`drug, voriconazole, increased oxycodone AUC and Cmax by 3.6 and 1.7 fold, respectively.
`Although clinical studies have not been conducted with other CYP3A4 inhibitors, the
`expected clinical results would be increased or prolonged opioid effects. If co-
`administration with OxyContin is necessary, caution is advised when initiating therapy
`with, currently taking, or discontinuing CYP450 inhibitors. Evaluate these patients at
`frequent intervals and consider dose adjustments until stable drug effects are achieved.
`[see Clinical Pharmacology (12.3)]
`
`Inducers of CYP3A4:
`
` 7
`
` DRUG INTERACTIONS
`
`(b) (4)
`
`(b) (4)
`
`(b) (4)
`
`

`

`
`
`
`
`
`
`
`
`
`CYP450 inducers, such as rifampin, carbamazepine, and phenytoin, may induce the
`metabolism of oxycodone and, therefore, may cause increased clearance of the drug
`which could lead to a decrease in oxycodone plasma concentrations, lack of efficacy or,
`possibly, development of abstinence syndrome in a patient who had developed physical
`dependence to oxycodone. A published study showed that the co-administration of
`rifampin, a drug metabolizing enzyme inducer, decreased oxycodone (oral) AUC and
`Cmax by 86% and 63% respectively. If co-administration with PERCODAN is
`necessary, caution is advised when initiating therapy with, currently taking, or
`discontinuing CYPP450 inducers. Evaluate these patients at frequent intervals and
`consider dose adjustments until stable drug effects are achieved.
`
`Inhibitors of CYP2D6:
`Oxycodone is metabolized in part to oxymorphone via cytochrome CYP2D6. While this
`pathway may be blocked by a variety of drugs (e.g., certain cardiovascular drugs
`including amiodarone and quinidine as well as polycyclic antidepressants), such blockade
`has not been shown to be of clinical significance during oxycodone treatment.
`
`12.3 Pharmacokinetics
`
`
`Drug-Drug Interactions
`
`
`Oxycodone is extensively metabolized by multiple metabolic pathways. CYP3A4 is the
`major enzyme involved in noroxycodone formation followed by CYP2B6, CYP2C9/19
`and CYP2D6. Drugs that inhibit CYP3A4 activity, such as macrolide antibiotics (e.g.,
`erythromycin), azole-antifungal agents (e.g., ketoconazole), and protease inhibitors (e.g.,
`ritonavir), may cause decreased clearance of oxycodone which could lead to an increase
`in oxycodone plasma concentrations. For example, a published study showed that the co-
`administration of the antifungal drug, voriconazole, increased oxycodone AUC and Cmax
`by 3.6 and 1.7 fold, respectively. Similarly, CYP450 inducers, such as rifampin,
`carbamazepine, and phenytoin, may induce the metabolism of oxycodone and, therefore,
`may cause increased clearance of the drug which could lead to a decrease in oxycodone
`plasma concentrations, lack of efficacy or, possibly, development of abstinence syndrome
`in a patient who had developed physical dependence to oxycodone. For example, a
`published study showed that the co-administration of rifampin, a drug metabolizing
`enzyme inducer, decreased oxycodone (oral) AUC and Cmax by 86% and 63%
`respectively.
`
`Oxymorphone is a minor metabolite, its formation is catalyzed primarily by CYP2D6 and
`to a small extent by CYP2C19. The formation of oxymorphone may be blocked by a
`
`(b) (4)
`
`

`

`variety of drugs (such as antipsychotics, beta blockers, antidepressants, etc.) that inhibit
`these enzymes. However, in a study involving ten subjects using quinidine, a known
`inhibitor of CYP2D6, the pharmacodynamic effects of oxycodone were unchanged. The
`genetic expression of CYP2D6 may have some influence in the pharmacokinetic
`properties of oxycodone.
`
`The in vitro drug-drug interaction studies with noroxymorphone using human liver
`microsomes showed no significant inhibition of CYP2D6 and CYP3A4 activities which
`suggests that noroxymorphone may not alter the metabolism of other drugs that

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