throbber
SAFETY ANALYSIS 56
`
`SECTION 8.0
`
`'
`
`SECTION 8.1
`
`EXPOSURE 56
`
`SECTION 8.2
`
`DEMOGRAPHICS: 57
`
`SECTION 8.3
`
`DEATHS so
`
`SECTION 8.4
`
`DISCONTINUA‘TIONS 63
`
`sECTION 8.5
`
`ALL ADVERSE EVENTS 74
`
`SECTION 8.6
`SECTION 8.6.1
`76
`
`SERIOUS ADVERSE EVENTS 76 .
`Serious adverse events for ABBOTT SPONSORED STUDIES (NON JAPANESE)
`
`SECTION 8.6.2
`SECTION 8.6.3
`
`serious adverse events for ABBOTT SPONSORED STUDIES (JAPANESE) 91
`serious adverse events IN NON ABBOTT SPONSORED STUDIES 91
`
`SECTION 8.7
`SECTION 8.7.]
`SECTION 8.7.2
`
`LABORATORY FINDINGS 94
`SERUM CHEMISTRY PARAMETERS 94
`vital signs and electocardiograms 95
`
`SECTION 8.8
`
`ADVERSE EVENTS AND PRECLINICAL STUDIES 98
`
`SECTION 8.9
`
`DOSE-RESPONSE DATA 98
`
`SECTION 8.10
`
`DRUG-DRUG INTERACTIONS 100
`
`SECTION 8.11
`
`DRUG-DEMOGRAPHIC INTERACTIONS 102
`
`SECTION 8.12
`
`LONG TERM ADVERSE EFFECTS 107
`
`SECTION 8.13 WITHDRAWAL EFFECTS _107
`
`“
`
`.
`
`SECTION 8.14
`
`120 SAFETY UPDATE 107
`
`SECTION 8.15
`
`SAFETY DISCUSSION 108
`
`SECTION 9.0
`
`_
`
`, DISOUSSION 110
`
`SECTION 16:0 IL’BECOMMENDATIONS 110
`
`APPENDIX I‘lil.
`
`_
`
`
`
`

`

`SECTION 1.0 MATERIALS USED IN REVIEW
`
`Table"'l
`
`MATERIALS UTILIZED IN REVIEW
`ITEM
`DATE
`
`
`
`Volumes 1,2,3
`
`Volumes 63-725
`“
`'
`
`
`
`18 December 1998
`
`5 Aril I999
`24 Ma 1999
`
`-»
`
`
`
`
`
`
`Introductory Information
`
`Proposed Labeling
`
`
`A- olication Summar
`
`Clinical Data
`Case Re -ort Forms
`
`
`Additional Information
`Additional Information
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`SECTION 2.0 BACKGROUND
`
`SECTION 2.1
`
`INDICATION
`
`Dexmedetomidine is an intravenous alpha-2 adrenoreceptor agonist indicated for sedation
`and- analgesia in an Intensive Care Unit setting.
`
`SECTION 2.2
`
`RELATED NDA’S AND IND’S
`
`All clinical studies were conducted
`
`
`
`— No previOus NDAs are applicable.
`
`SECTION 2.3
`
`PROPOSED DIRECTIONS FOR USE
`
`Dexmedetomidine is proposedfor adults 18 years and older in an Intensive Care Setting
`who require sedation or potentiation' of analgesia for up to 24 hours. The drug will be
`administered intravenously. Dosing is initiated with a loading dose of l pig/kg over 10
`minutes followed by a maintenance dose'o£0.2-O._7 ug/kg/hour; dosing is adjusted to
`achieve the desired-‘Ievel of"sedation. The total daily dose will not‘exCeed 20 ug/kg with a
`daily steady state plasma concentration exposure of' less than 3.0 ng/ml.
`-
`
`SECTION 2.4.
`
`FOREIGN MARKETING
`
`Dexmedetomidine is not marketed anywhere in the world.
`
`

`

`
`
`SECTION 3.0 CHEMISTRY
`
`
`
`Sponsor states the solution should be stored at room temperature (59 to 86 degree F).
`
`SECTION 4.0 ANIMAL PHARMACOLOGY/TOXICOLOGY
`
`The following is a summary of the pharmacology/toxiCOlogy provided by the sponsor:
`
`“Alpha-2 adrenoceptor agonists have been in clinical use since the mid 19605 with the
`introduction of clonidine, the archetypal alpha-2 adrenoceptor agonist. Although
`clonidine has been used as an antihypertensive drug, it is also used as an adjunctive
`sedative in the intensive care setting.
`'
`
`Alpha-2 adreneeeptor activation is known to result in a variety of responses from several
`organs and tissues. A prominent effect is reduction in sympathetic nervous activity.
`Activation of presyriaptic alpha-2 adrenoceptors located in sympathetic nerve endings
`inhibits the rEIEase‘bf the neurotransmitter noradrenaline. Activation of postsynaptic
`alpha-2 adrenoceptors in the CNS leads to inhibition of sympathetic activity, causing
`decreases in blood pressure and heart rate, sedation, and relief of anxiety. Activation of
`alpha-2 adrenoceptors at the spinal cord results in analgesia. Peripheral alpha-2
`adrenoceptors in blood vessels mediate vascular smooth muscle contraction.
`‘
`
`Alpha-2 adrenoceptors agonists can decrease stress induced ACI‘H release and hence
`cortisol synthesis by a direct effect on the brain. Alpha-2 adrenergic agonists can inhibit
`insulin release by actions on pancreatic islet cells and have been shown to stimulate
`
`

`

`
`
`growth hormone. Subcutaneous administration of Dexmedetornidine increased blood
`glucose, decreased insulinsecretion, and inhibited lipolysis. Alpha-2 adrenoceptor
`agonists decrease circulating norepinephrine and epinephrine by central and peripheral
`mechanisms.
`
`Dexmedetornidine is a specific alpha-2 adrenoceptor agonist as shown by both receptor
`binding and functional studies. Dexmedetornidine has_very low affinity for alpha-l
`adrenoceptors (1300 times less than for alpha-2 adrenoceptors in the rat membrane
`model) and negligible affinity for other receptors, including beta adrenergic, muscarinic,
`dopamine, serotonin, mu- and delta opiate, GABA, andbenzodiazepine receptors.
`Dexmedetornidine is a lipophilic compound which is rapidly and extensively distributed
`to tissues and rapidly eliminated.
`"
`
`Dexmedetornidine is an alpha-2 sedative in various animal species including the rat, dog,
`rabbit, and mouse producing dose dependent sedation/hypnosis when administered either
`intracerebroventricularly, subcutaneously, intraperitoneally, _or intravenously. The
`sedative effect is biphasic: lower (IO-300 ug/kg) doses cause maximal sedation and
`higher doses (2 1000 ug/kg) result in a reversal of the Dexmedetornidine sedative effect.
`The reversal of the sedative effect seen at higher doses of Dexmedetomidine is
`hypothesized to be due to the activation of alpha-l adrenoceptors by Dexmedetornidine,
`as the reversal-could also be blocked by the alpha-l antagonist prazosin. At low doses
`(< 3 ug/kg) Dexmedetornidine is anxiolytic in mice and rats. It acts synergistically with
`midazolam, diazepam, or fentanyl to induce sedation/hypnosis and has potent volatile
`anesthetic sparing properties which are mediated via central alpha-2 adrenoceptors.
`
`Dexmedetomidine administered both spinally and peripherally to rats, dogs, mice
`monkeys, and sheep produces dose dependent analgesia which is more potent than that
`caused by clonidine, ST-9l, xylazine, epinephrine, or‘norepinephrine. The analgesia
`effect in animals lasts 1 to 8 hours depending on the route of administration. The
`analgesic effects of Dexmedetomidine are mediated by alpha~2 adrenoceptors.
`
`Lower doses of Dexmedetornidine cause reduction in blood pressure and heart rate
`through a central effect whereas higher doses of Dexmedetomidine result in peripheral
`alpha-1 adrenoceptor activation and resultant higher blood pressure. The initial
`hypertensive efietof Dexmedetornidine seen with IV bolus injections is reduced with a
`slower rate of infusion. At doses causing increases in mean arterial pressure,
`Dexmedetomidinejeduces heart rate and cardiac output in a dose dependent manner.
`Dexmedetornidine'has no direct depressant effect on the myocardium except at a very
`high supra-clinical concentration. Through its sympatholytic action, Dexmedetornidine
`depresses cardiac function and contractility; the effect on cardiac function and
`contractility are dose dependent.
`
`Dexmedetomidine produces no significant effect on respiratory function except for mild
`respiratory depression at high supra-clinical doses.
`
`

`

`
`
`
`
`Other effects of Dexmedetomidine in animals include reduced seizure thresholds,
`modulation of body temperature, and induction of hypothermia. Dexmedetomidine acts in
`an additive manner with opioids in producing analgesia and counteracts opioid induced
`muscle rigidity.
`
`Animal deaths have been reported only after the administration of doses of
`Dexmedetomidine exceeding the LDSO. All adverse effects are extensions of alpha-2
`adrenergic activity.”
`
`SECTION 5.0 “SUMMARY OF HUMAN PHARMOKINETICS
`
`The pharmacokinetic and pharmacodynamic profiles of Dexmedetomidine were based on
`data from lft__studies. The sponsor has summarized the human’pharmacokinetic and
`bioavailability data as follows:
`
`“Dexmedetomidine is extensively metabolized in humans. In a radiolabeled
`Dexmedetomidine study, there was virtually no penetration of radioactivity into the
`cellular fraction. The major circulating metabolites are the N-glucuronides of
`Dexmedetomidine. Other minor metabolites include the carboxy (COOH), N-methylated
`(N-meth), the glucuronide conjugate of hydroxylated Dexmedetomidine, and additional
`unidentified minor metabolites.
`
`The pharmacokinetics of Dexmedetomidine are biphasic with rapid distribution
`(t l/2 0t 2 6 min) and a mean terminal half life of approximately 2.0 to 2.5 hours.
`Following the loading infusion, venous plasma concentrations rise rapidly. Due to the
`rapid distribution pharmacokinetics, concentrations drop quickly when the loading
`infusion stops, after which the combined effects of the loading and maintenance infusions
`hold plasma concentrations stable until the infusion is terminated. Dexmedetomidine is
`almost exclusively eliminated by metabolism; 95% of a radioactive dose is excreted as
`conjugates in the urine, and the remainder in the feces.
`
`The following were measured PK parameters in healthy human subjects:
`
`Table 2
`
`
`:Wnfieiimedetomidine Pharmacokinetic Parameters
`
`Parameter
`
`
`
`
`
`
`143.9 1155
`
`AUC (ng-h/g)
`
`Clearance (Uhour>
`
`
`
`
`
`
`
`Modified Sponsor’s Table 3 Vol 8/10-1-69
`
`
`
`

`

`8
`
`Dexmedetomidine is 93.7% bound to plasma proteins. There is no difference in binding
`due to gender, and it does not differ in normal subjects or subjects with mild, moderate,
`or severe renal impairment. Subjects with hepatic impairment have protein binding that is
`82-88% of the protein binding of normal subjects depending on the severity of the
`impairment.
`
`Mean arterial pressure, systolic blood pressure, and diastolic blood pressure showed a
`biphasic response with initial decreases at the lowest concentrations, followed by a return
`to baseline and increases over the mean baseline level when the plasma concentration of
`Dexmedetomidine was greater than 3.2 ng/ml. The mean heart rate decreased
`progressively until. the mean plasma concentration of Dexmedetomidine was 3.2-5.1
`ng/ml; at this concentration heart rate reached a plateau and did not decline further.
`Cardiac output decreased approximately 20-30% at Dexmedetomidine plasma ~
`concentrations of 2-4 ng/ml in most subjects. At concentrations above 4 ng/ml, no further
`decrease incardiac output was observed. There was no evidence of respiratory
`depression.
`
`Special Populations:
`
`Renal Failure: During a 2 day study period, six severely renally impaired subjects
`(creatinine clearance < 3 ml/min) received a single 10 minute intravenous infusion of
`Dexmedetomidine 0.6 ttg/kg; they were compared to healthy subjects. Pharmacokinetics,
`sedation, heart rate, blood pressure, oxygen saturation,'respiratory rate, and safety were
`assessed. Blood and urine samples were collected for assay of Dexmedetomidine and
`Dexmedetomidine metabolites. The collection of the samples occurred prior to closing,
`during the infusion, and at protocol specified time points up to 24 hours post-dosing.
`There were no statistically significant differences between the two groups for any of the
`pharmacokinetic parameters. There were no clinically relevant differences between
`healthy subjects and renally impaired subjects. The renal subjects were slightly more
`sedated than control subjects.
`‘
`
`Hepatic Impairment: During a 3 day study, 18 subjects with hepatic impairment received
`a single 10 minute intravenous infusion of Dexmedetomidine Doug/kg; they were
`compared to healthy subjects. The objective of the study was to evaluate the effect of
`various degreesofhepatic impairment on the pharmacokinetics of Dexmedetomidine. In
`addition, sedation, heart rate, blood pressure, oxygen saturation, respiratory rate, and
`safety were assessed. The hepatic subjects contained groups with mild, moderate, and
`severe hepatic: fail-tire. Dexmedetomidine clearance values for'subjects with mild,
`moderate, and severe hepatic impairment were only 74%, 64%, and 53%, respectively of
`those observed in the normal healthy subjects. Subjects with hepatic impairment appeared
`to have no clinically relevant different effects on hemodynamic or respiratory parameters.
`Sedation may have been greater in the hepatically impaired group. Therefore, the dose of
`Dexmedetomidine may need to be reduced in subjects with hepatic impairment.
`
`Drug Interaction: 6 studies in humans were performed to evaluate possible interactions
`between Dexmedetomidine and other drugs. 5 of these studies involved pharmacokinetic
`
`
`
`

`

`
`
`analysis. The agents studies were isoflurane, rnidazolam, alfentanil, propofol,
`rocuronium, and esmolol (no PK analyses were performed with esmolol). Except for
`alfentanil, no pharmacokinetic interactions were observed. PK effects by
`Dexmedetornidine on alfentanil were observed along with enhancement of alfentanil’s
`analgesic effects.
`
`Age: The influence of age on the pharmacokinctics ofDexmedetornidine in healthy
`subjects was evaluated by administration of 0.6 ug/kg Dexmedetornidine given as a 10
`minute intravenous infusion. Subjects were studied up to 24 hours following infusion.
`Age groupings varied from 18 years to > 65 years. Dexmedetomidine pharmacokinetics
`were not different between young, middle aged, and elderly subjects nor between male
`' and female subjects when differences in body weight were accounted for. Therefore the
`same Dexmedetornidine infusion is expected to produce the same Dexmedetom‘idihe
`profile, and the elimination characteristics are expected to be the same irrespective of age
`or gender.”— -
`
`SECTION 6.0 DESCRIPTION OF CLINICAL DATA
`SOURCES
`
`SECTION 6.1
`
`DEVELOPMENT
`
`Sponsor presents the following information on development of Dexmedetomidine:
`
`“me05 Group, Finland began research involving alpha-2 adrenoceptor agonists and
`antagonists in the late 19705. One of the first agents Was detomidine which has-a higher
`affinity for the alpha-2 adrenoceptor than clonidine. Detomidine was developed for use as
`a sedative/analgesic in horses and cattle and it was first registered for marketing in
`Finland in 1983. Medetomidine, a more specific and selective alpha-2 adrenoceptor
`agonist than clonidine, was launched in 1987 in Finland and the Scandinavian countries
`for use as a veterinary sedative/analgesic in dogs and cats; it was approved for use in
`animals in the US in 1997. The sedative and anesthetic pharmacological activity of
`medetomidineresides predominantly in the dextro enantiomer, Dexmedetomidine, which
`was first synthesized by Farmos group in 1986.
`
`Clinical worlEin the US began with the submission of an Investigational New Drug
`Application in 1989. Clinical investigation included many potential applications of
`Dexmedetornidine from preoperative through postOperative periods. In 1990 Farmos
`
`merged with Orion Corp. _‘
`'
`"
`'
`‘
`'
`'
`ge form of Dexmedetornidine
`for clinical use to Abbott Laboratories in 1994."
`
`SECTION 6.2_
`
`OVERVIEW OF CLINICAL STUDIES
`
`
`
`
`
`

`

`10'
`
`- conducted a total of 56 clinical trials in which a total of 1527 subjects/patients
`were exposed to Dexmedetomidine. The
`‘\—-——;—~ ‘1 studies evaluated the use of
`Dexmedetomidine in the perioperative setting and utilized various modes of
`administration includingrapid IV infusion, continuous IV infusion, and IM injection.
`Transdermal and oral administration of Dexmedetomidine were also studied in a limited
`capacity.
`
`Abbott Laboratories initiated its own clinical program to evaluate Dexmedetomidine in
`the perioperative setting using IV infusion administration. A total of 21 studies ( 13 Phase
`I and 8 Phase II/III) have been completed in the US, Canada,, and Europe in support of
`the perioperative program, during which a total of 230 subjects were closed and 767
`' patients received Dexmedetomidine. 13 of the 21 studies were Phase I trials assessing the
`pharmacokinetic/pharmacodynamic properties of Dexmedetomidine, the safety profile.
`the potential for drug interactions, and its use in special populations. The remaining 8
`studies were Phase II/III trials evaluating the use of Dexmedetomidine as an anesthetic
`adjunct in patients undergoing major surgery and electroconvulsive therapy. The effect
`on minimum alveolar concentration of an inhalation anesthetic was also studied. 4 studies
`were conducted in Europe and Canada by Abbot Labs to evaluate Dexmedetomidine’s
`potential as a sedative and analgesic agent for patients in the intensive care setting. A
`total of 631 subjects/patients were dosed/treated with Dexmedetomidine in these studies.
`
`SECTION 6.3 DEMOGRAPHICS
`
`See Section 8.2
`
`SECTION 6.4
`
`EXTENT OF EXPOSURE
`
`See Section 8.1
`
`SECTION-LP EFFICACY FINDINGS
`
`SECTION'7i1‘: OVERVIEW OF CLINICAL EFFICACY STUDIES:
`
`The Sponsor identified two Phase III studies that were conducted in support of this NDA
`application: W97-245 and W97-246. Sponsor also identified one Phase II study
`supporting the application, W97-249. Study W97-249 randomized 12 patients in one
`center and was not reviewed for findings of efficacy.
`'
`
`
`
`
`
`

`

`11
`
`SECTION 7.2
`
`SUMMARY OF STUDIES PERTINENT TO
`EFFICACY
`
`SECTION 7.2.1
`
`STUDY W97-245
`
`SECTION 7.2.1.1 5 PROTOCOL REVIEW SUMMARY
`
`TITLE: 'A Phase III,,‘Multi-Center, Randomized, Placebo-Controlled, Double-Blind
`Study Evaluating the Safety and Efficacy of Dexmedetomidine When
`Compared to Placebo, With Midazolam, in ICU Sedation in Post-Operative
`Patients
`
`OBJECTIVES:
`
`Primary:
`
`The primary objective of this two-part, Phase III study was to evaluate the
`efficacy and safety of Dexmedetomidine in patients requiring ventilation,
`sedation, and intensive care following surgery. Dexmedetomidine was to be
`administered as clinically indicated according to Ramsay sedation scores
`[see Appendix 1 for description of Ramsay sedation score]; the goal was to
`achieve Ramsay scores of 2 3, as clinically indicated. The primary efficacy
`variable for this study was the total dose (mg) of midazolam required in
`addition to the study drug to achieve adequate sedation (as clinically
`determined by the Ramsay sedation scale) during intubation.
`
`Secondary: Secondary variables included total dose of midazolam (mg) administered
`during study drug infusion, use of morphine for pain, as assessed by total
`dose used with Dexmedetomidine as compared to placebo; use of
`paracetamol for pain after extubation. as assessed by total dose used with
`Dexmedetomidine as compared to placebo, and time to extubation. as
`measured by time to arrival in ICU until time to extubation.
`
`:. as.
`
`STUDY DESIGN: .
`This was a two part study in postoperative patients requiring a minimum of 6 hours
`ventilation and sedation in ICU. Part I was open-label to allow the investigator to become
`more familiar with the observed clinical effects of Dexmedetomidine prior to starting the
`double-blind portion of the study. Part II was double blind, randomized, and placebo-
`controlled.
`.
`
`Patients were to be screened within 7 days prior to receiving study drug. Screening was to
`include a complete medical history and physical examination, laboratory assessments,
`and 12-lead electrocardiogram. Study drug administration was to be initiated as soon as
`possible after arrival in the ICU but not later than 1 hour after admission to the ICU. If
`
`
`
`

`

`_
`
`12
`
`possible, study drug was to be started prior to the patient’s awakening in the ICU or
`requiring any other medication for sedation. If a patient required sedation post-surgery
`and prior to the start of study drug, midazolam (l-mg bolus) could be given as required.
`Study drug infusion was to be continued for 6 hours post extubation. The investigator
`_
`could have continued the infusion at his/her discretion for a maximum of 24 hours.
`Patients were to be observed for a 24-hour period after the end of the study drug infusion.
`
`Part I of the study was to include up to 4 patients per site. Patients were to be
`administered a loading dose of 6.0 mcg/kg/h of Dexmedetomidine over a 10 minute
`period, followed by a maintenance infusion of 0.4 mcg/kg/h. Following the initial
`.:
`maintenance infusion, the rate was to be adjustedif clinically necessary, in increments of
`0.1 mcg/kg/h or higher. The infusion rate was to be maintained between a range of 0.2 to
`0.7 mcg/kg/h to achieve and maintain a Ramsay sedation score of 3 or higher (as '
`clinically appropriate for the patient’s needs). Following exiubation, the infusion rate .
`could havebeen adjusted to achieve a Ramsay sedation score of 2 and above (as
`clinically appropriate).
`'
`
`In Part II of the study (double blind, randomized, placebo controlled), approximately 300
`patients were to be randomized to one of two treatment groups: Dexmedetomidine or
`placebo with additional doses of midazolam for sedation administered as clinically
`indicated. Patients were administered a two stage infusion consisting of a 10 minute
`loading dose of 6.0 mcg/kg/h of Dexmedetomidine or placebo followed by a maintenance
`infusion of 0.4 mcg/kg/h. Following the initial maintenance infusion, the rate could be
`adjusted in increments of 0.1 mcg/kg/h or higher, and was to be maintained in the range
`of 0.2 to 0.7 mcg/kg/h as clinically deemed necessary to achieve and maintain a Ramsay
`sedation score of 3 or higher as clinically appropriate. Following extubation, the infusion
`rate was to be adjusted to achieve a Ramsay sedation score of 2 or above as clinically
`appropriate.
`"
`
`INCLUSION CRITERIA:
`To be included in the study, patients were to have satisfied all of the following inclusion
`criteria:
`
`0
`
`Signed and dated the Informed Consent after the study had been fully explained or
`had a legally acceptable representative sign and date the Informed Consent.
`0 Required sedation for ventilation and intensive care for a minimum of 6 hours
`following surgery
`0 Male or female; age 18 and over (in Austria, age 19 or older)
`0
`If female and of child bearing potential, was not pregnant (confirmed by negative
`pregnancy test) and not lactating.
`'
`-
`
`EXCLUSION CRITERIA:
`
`Patients were not eligible for the study if they met any of the following criteria:
`0 Had serious central nervous system trauma.
`0 Had undergone or required intracranial surgery during current hospitalization.
`
`

`

`13
`
`0 Required the use of neuromuscular blocking agents during the study period, except
`for the insertion of the endotracheal tube.
`0 Required epidural or spinal analgesia during the ICU stay.
`0
`In whom opiates or benzodiazepines were contraindicated or had known or suspected
`serious allergy to any medication that might have been administered during the course
`of the study.
`0 Was grossly obese (estimated body weight was greater than 50% above ideal body
`weight)
`0 Was currently hospitalized for drug overdose
`r -In whom alpha-2 antagonists or alpha-2 agonists were contraindicated
`0‘ Was currently being treated or had been treated within the last 30 days with alpha-2
`agonists or antagonists
`_
`.
`0 Had participated in a trial with any experimental drug within 30 days prior to
`admission to the ICU
`' ‘
`
`0 Was terminally ill, whose life duration expectancy was no more than or around 24
`hours.
`
`Was considered unable to undergo any procedure required by the protocol
`Had demonstrated tolerance to standard sedating medications
`Had previously received Dexmedetomidine
`Had unstable or uncontrolled diabetes
`Had excessive bleeding which was likely to require resurgery
`. Had received midazolam for maintenance of anesthesia
`Has clinically significant arrhythmia or any other cardiac condition or factor which,
`in the investigator’s opinion, might have increased the risk to the patient or precluded
`obtaining satisfactory data.
`'
`
`REMOVAL OF PATIENTS FROM THERAPY ASSESSMENT:
`A patient was to be withdrawn from the study immediately if any of the following
`occurred:
`'
`
`0 Due to an adverse event, the investigator decided that discontinuation was in the
`patient’s best interest.
`0 The patient requested withdrawal from the study.
`0
`Patients requiring reoperation
`0 A changeeceurred in the patient’s status such that exclusion criteria became part of
`the patient profile.
`
`Patients who: withdrew from the study were not to be replaced. Those patients
`withdrawn fi'om the study due to an adverse event were to have all events documented
`and followed to a satisfactory resolution. Patients who were withdrawn from the study
`for any reason during study drug administration were required to have all final
`evaluation procedures completed.
`
`DOSING SCHEDULE:
`
`Dexmedetomidine or placebo was to be administered as a two-stage infusion: a 10
`minute loading dose followed by a maintenance infusion using standard syringe pump
`
`

`

`l4
`
`and IV administration sets. Study drug was never to be administered directly‘into the
`pulmonary artery. Study drug was to be initiated as soon as possible after arrival in the
`ICU but no later than 1 hour after admission to ICU. If possible, study drug was to be
`started prior to the patient’s awakening in ICU or requiring any other medication for
`sedation. If a patient required sedation post surgery and prior to start of study drug,
`additional doses of rnidazolam for sedation could have been given as required. Drug
`administration consisted of a 10 minute loading dose of 6.0 mcg/kg/h followed by a
`maintenance infusion of 0.4 mcg/kg/h. The 6 mcg/kg/h loading dose was chosen to
`' achieve a Dexmedetomidine plasma concentration of approximately 1.5 rig/mg as a
`result of experience gained in Phase I studies and resultant PK modeling. Clinical
`.
`effects of sedation should have been observed within 15 minutes of the start of the study
`' drug. The infusion rate could have been adjusted in increments of 0.1 mcg/kg/h or
`higher, and should have been maintained in the range of 0.2:0.7 mcg/kg/h as clinically
`deemed necessary to achieve and maintain a Ramsay sedation score of at least 3 as
`clinically appropriate. Following extubation, the infusion rate could have been adjusted
`to achieve a Ramsay sedation score of 2 and above as clinically appropriate.
`
`During study drug administration, rescue medication was limited to rnidazolam for
`sedation and morphine for pain as required. After extubation, paracetamol use was to be
`permitted as clinically indicated. During the 10 minute loading dose 1 mg bolus dose of
`midazolam was allowed if necessary. The maintenance dose of Dexmedetomidine or
`placebo was to be adjusted prior to any administration of additional rnidazolam. Study
`drug infusion was to be continued for 6 hours post extubation. The investigator may
`have continued the infusion at his/her discretion for a total of 24 hours total drug
`infusion.
`
`In the ICU, rnidazolam for sedation was to be administered following an increase in
`study drug infusion. Prior to the administration of rnidazolam, patients were to be
`assessed for sedation using the Ramsay sedation scale. The Ramsay assessment was to
`be performed prior to and 10 minutes after every rate change in study drug
`administration or administration of any rnidazolam. Pain was assessed either by direct
`corrununication with the patient or by autonomic signs (sweating, tachycardia,
`hypertension). Sponsor recommended that initial doses of rnidazolam be administered as
`a bolus in doses of 0.02 mg/kg. If, in addition to increasing the infusion rate of study
`drug, the patientreceived 3 boluses of rnidazolam within any 2 hour (during study drug
`infusion), further midazolam, if necessary, may have been administered at a continuous
`infusion rate of 0_.Ol-0.0Z mg/kg/h. Morphine may have been administered for pain in
`increments'o’f 2-mg IV boluses. Prior to the administration of morphine, the patient was
`to be assessed for pain.
`'
`_
`
`Standard ICU monitoring protocols were to be employed. All patients were to be
`ventilated to maintain PaC02 and Pa02 tensions as determined by the investigator.
`Minimum ventilatory support was to be utilized. Patients were to be weaned from the
`ventilator and extubated only if the investigator deemed it appropriate and after meeting
`the following criteria:
`0
`Patient was aWake or arousable, neurologically intact, cooperative, and comfortable.
`
`
`
`

`

`
`
`15
`
`0
`
`0
`
`Patient had an Fi02 value 5 0.4, PEEP < 5 cm H20, and pressure support 5 10cm
`H20.
`‘
`
`Patient had the following lung mechanics: minute ventilation expired > 4 Urnin but
`< 15 Umin, tidal volume > 5 mng and spontaneous respiratory rate <25/rnin.
`
`The following drugs were not to be allowed during study drug infusion:
`o
`Sedating agents Other than midazolam; analgesic agents other than morphine (after
`extubation, use of paracetamol was permitted as clinically indicated).
`0 Neuromuscular blocking agents except for the insertion of the endotracheal tube.
`0" 'Epidural or spinal analgesic agents.
`.
`. 6 Any drugs contraindicated with the use of Dexmedetomidine, midazolam. or
`morphine.
`-
`o Alpha-2 agonist/antagonist.
`
`SECTION 7.2.1.2
`
`STATISTICAL ANALYSIS
`
`Only patients from the randomized, double-blind, placebo-controlled part of the study
`(Part II) were to be included in the efficacy analyses. A patient was required to satisfy the
`following evaluability criteria in order to be included in the evaluable subset:
`o The patient received study drug for at least 6 hours,.unless the patient was
`I prematurely discontinued by the investigator due to an adverse event.
`0 The patient received none of the following medications during study drug
`administration: sedating agents other than midazolam, analgesic agents other than
`morphine or paracetamol, neuromuscular blocking agents except for insertion of the
`endotracheal tube, epidural or spinal analgesic/anesthetic agents, any drugs .
`contraindicated with the use of midazolam, Dexmedetomidine, or morphine, or other
`prohibited medications
`.
`o The patient received only morphine or paracetamol for 'pain management.
`0 The patient was intubated for at least 6 hours.
`
`A patient was included in the intent-to-treat subset if he or she was randomized and
`required intensivegare and sedation following surgery. Patients in Part I and H of the
`study were included in the safety subset if he/she received any study drug.
`
`EFFICACY ANAEYSES:
`The primary efficacy analysis was based on the intent-to-treat subset of patients._A
`second set of efficacy analyses was completed on the evaluable subset.
`
`Primary Efficacy Analysis
`
`The primary efficacy variable in this study was the total dose (mg) of midazolam during
`intubation received as rescue medication for sedation during the period of study drug
`administration. The total dose was summarized by the number of patients in the following
`three total dose categories: no midazolam (0 mg); a subtherapeutic dose over time (>0 mg
`
`
`
`

`

`16
`
`to 4 mg); and a therapeutic dose (>4 mg). Differences in the distributions of the
`proportion of patients in each category between the dexmedetomidine treatment group
`and theplacebo treatment group were tested with a chi-square statistic. Center differences
`were also explored. The total dose was also summarized by N, mean, standard error of
`the mean (SEM), minimum, median, and maximum The treatment groups were
`compared using an analysis of variance (ANOVA) with treatment, center and treatment-
`by-center interactionincluded in the model.
`
`.
`
`[Reviewer Note: The final primary eflicacy analysis submitted in this application is '
`difi’erentfrom the sponsor’s proposal in the original protocol. None of the amendments to
`this study reflect the analysis that was performed. ‘At a meeting with the sponsor at the
`conclusion of the Phase II studies, Dr. Thomas Permutt (the_reviewing statistician)
`suggested that the capability of Dexmedetomidine to provide sedation would be more
`convincinglydemonstrated by an analysis ofhow many patients needed any rescue
`medication rather than by measuring the‘amount of rescue medication utilized by both
`placebo and Dexmedetomidine patient groups. Consequently, the sponsor was '
`encouraged to incorporate calculations of the number ofpatients receiving any amount
`of midazolam in the primary efficacy analysis. ]
`
`Secondary Efficacy Analyses
`
`The following are secondary efficacy variables in the study:
`
`0 Total dose of midazolam during study drug administration:
`
`The total dose of midazolam (mg) administered during study drug infusion was
`calculated. The total dose was divided by the length of infusion to determine the total
`dosing rate during infusion and was expressed as mg/h. The length of infusion was
`defined as the difference between the time ofthe start of study drug and the end of
`study drug infusion. The total dosing rate was summarized by N, mean, SEM,
`median, minimum. and maximum. The treatment groups were compared using an
`ANOVA with treatment, center, and treatment-by-center interaction included in the
`model.
`
`"L;
`
`.3.
`
`o
`
`The total dose of morphine during study drug administration:
`
`Analysis oftot'al dose of morphine during study drug administration was run on three
`populations of patients. The first population consisted of patients who did not receive
`any midazolam during intubation. The second population consisted of patients with a
`subtherapeutic total dose over time of midazolam during intubatio

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