throbber
Feature Review Articles
`
`Dexmedetomidine: Applications in pediatric critical care and
`pediatric anesthesiology
`
`Joseph D. Tobias, MD
`
`Objective: To provide a general descriptive account of the
`end-organ effects of dexmedetomidine and to provide an evi-
`dence-based review of the literature regarding its use in infants
`and children.
`Data Source: A computerized bibliographic search of the lit-
`erature regarding dexmedetomidine.
`Main Results: The end-organ effects of dexmedetomidine have
`been well studied in animal and adult human models. Adverse
`cardiovascular effects include occasional episodes of bradycardia
`with rare reports of sinus pause or cardiac arrest. Hypotension
`has also been reported as well as hypertension, the latter thought
`to be due to peripheral ␣2B agonism with peripheral vasoconstric-
`tion. Although dexmedetomidine has no direct effects on myocar-
`dial function, decreased cardiac output may result from changes
`in heart rate or increases in afterload. There are somewhat
`conflicting reports in the literature regarding its effects on ven-
`tilatory function, with some studies (both human and animal)
`suggesting a mild degree of respiratory depression, decreased
`minute ventilation, and decreased response to CO2 challenge
`
`whereas others demonstrate no effect. The central nervous sys-
`tem effects include sedation and analgesia with prevention of
`recall and memory at higher doses. Dexmedetomidine may also
`provide some neuroprotective activity during periods of ischemia.
`Applications in infants and children have included sedation during
`mechanical ventilation, prevention of emergence agitation follow-
`ing general anesthesia, provision of procedural sedation, and the
`prevention of withdrawal following the prolonged administration
`of opioids and benzodiazepines.
`Conclusions: The literature contains reports of the use of
`dexmedetomidine in approximately 800 pediatric patients. Given
`its favorable sedative and anxiolytic properties combined with its
`limited effects on hemodynamic and respiratory function, there
`is growing interest in and reports of its use in the pediatric
`population in various clinical scenarios. (Pediatr Crit Care Med
`2007; 8:115–131)
`KEY WORDS: dexmedetomidine; ␣2-adrenergic agonist; opioid
`tolerance and withdrawal; emergence delirium; procedural-
`sedation
`
`D exmedetomidine (Precedex,
`
`Hospira Worldwide, Lake For-
`est, IL) is the pharmacologi-
`cally active dextro-isomer of
`medetomidine. It exerts its physiologic ef-
`fects via ␣2-adrenergic receptors. The ␣2-
`adrenergic agonists are subclassified into
`three groups: imidazolines, phenylethyl-
`amines, and oxalozepines. Dexmedetomi-
`dine and clonidine are members of the
`imidazole subclass, which exhibits a high
`ratio of specificity for the ␣2 vs. the ␣1
`receptor (Fig. 1). Clonidine exhibits an
`␣2:␣1 specificity ratio of 200:1 whereas
`that of dexmedetomidine is 1600:1,
`thereby making it a complete agonist at
`the ␣2-adrenergic receptor (1). Dexme-
`detomidine has a short half-life (2–3 hrs
`
`From the Departments of Anesthesiology and Pe-
`diatrics, University of Missouri, Columbia, MO.
`The author has consulted for and received hono-
`raria/speaking fees from Hospira.
`Copyright © 2007 by the Society of Critical Care
`Medicine and the World Federation of Pediatric Inten-
`sive and Critical Care Societies
`DOI: 10.1097/01.PCC.0000257100.31779.41
`
`Pediatr Crit Care Med 2007 Vol. 8, No. 2
`
`vs. 12–24 hrs for clonidine) and is com-
`mercially available for intravenous ad-
`ministration. An epidural clonidine for-
`mulation, although not marketed for
`intravenous administration, has been
`used for this purpose in clinical practice
`without consequences. Dexmedetomi-
`dine’s end-organ effects are mediated via
`postsynaptic ␣2-adrenergic receptors and
`subsequent activation of a pertussis tox-
`in-sensitive guanine nucleotide regula-
`tory protein (G protein) (2), which results
`in inhibitory feedback and decreased ac-
`tivity of adenylyl cyclase (3). A reduction
`of intracellular cyclic adenosine mono-
`phosphate and intracellular cyclic adeno-
`sine monophosphate-dependent protein
`kinase activity results in the dephosphor-
`ylation of ion channels (4). Alterations in
`ion channel function, ion translocation,
`and membrane conductance lead to de-
`creased neuronal activation and the clin-
`ical effects of sedation and anxiolysis (5).
`Centrally acting ␣2-adrenergic agonists
`also activate receptors in the medullary
`vasomotor center, reducing norepineph-
`rine with a resultant central sympatho-
`
`lytic effect leading to decreased heart rate
`(HR) and blood pressure (BP). As the cen-
`tral presynaptic ␣2A-adrenergic receptor
`is a negative feedback receptor, agonists
`at this receptor result in decreased cate-
`cholamine release from the nerve termi-
`nal. Central nervous system stimulation
`of parasympathetic outflow and inhibi-
`tion of sympathetic outflow from the lo-
`cus ceruleus in the brainstem play a
`prominent role in the sedation and anxi-
`olysis produced by these agents. De-
`creased noradrenergic output from the
`locus ceruleus allows for increased firing
`of inhibitory neurons, most importantly
`the ␥-aminobutyric acid system (6 – 8).
`Primary analgesic effects and potentia-
`tion of opioid-induced analgesia result
`from the activation of ␣2-adrenergic re-
`ceptors in the dorsal horn of the spinal
`cord and the inhibition of substance P
`release. These interactions with central
`nervous system and spinal cord ␣2-
`adrenergic receptors mediate dexmedeto-
`midine’s primary physiologic effects in-
`cluding sedation, anxiolysis, analgesia, a
`decrease of the minimum alveolar con-
`
`115
`
`Petition for Inter Partes Review of US 8,455,527
`Amneal Pharmaceuticals LLC – Exhibit 1061 – Page 115
`
`

`

`Figure 1. Representation of the chemical struc-
`ture of clonidine and dexmedetomidine, ␣2-
`adrenergic agonists of the imidazole subclass,
`which exhibit a high ratio of specificity for the ␣2
`vs. the ␣1 receptor.
`
`inhalational anesthetic
`centration of
`agents, decreased renin and vasopressin
`levels leading to diuresis, blunting of the
`sympathetic nervous system, and lower-
`ing of HR and BP (Fig. 2) (9, 10).
`Currently, dexmedetomidine’s only
`Food and Drug Administration (FDA)-
`approved indication is the provision of
`short-term sedation (⬍24 hrs) in adult
`patients in the intensive care unit (ICU)
`setting who are initially intubated and
`receiving mechanical ventilation (11). It
`is available in a water-soluble solution
`without the addition of lipid or propylene
`glycol and is not associated with pain
`following intravenous administration.
`There are no active or toxic metabolites.
`Given its favorable physiologic effects
`combined with a limited adverse effect
`profile reported to date, there is increas-
`ing use of this agent in the pediatric
`population. This article reviews the basic
`pharmacology of dexmedetomidine,
`its
`end-organ effects and adverse effect pro-
`file, and reports from the literature re-
`garding its use in various clinical scenar-
`ios in infants and children.
`
`PHARMACOKINETICS
`
`In healthy adult volunteers, dexme-
`detomidine’s pharmacokinetic profile in-
`
`Figure 2. The physiologic end-organ effects of dexmedetomidine.
`
`cludes a rapid distribution phase (distri-
`bution half-life of 6 mins), an elimination
`half-life of 2 hrs, and a steady-state vol-
`ume of distribution of 118 L (12). In the
`dosing range of 0.2– 0.7 ␮g/kg/hr deliv-
`ered via continuous intravenous infusion
`for up to 24 hrs, the pharmacokinetics
`are linear. Dexmedetomidine is 94% pro-
`tein bound to serum albumin and ␣1-
`glycoprotein. It undergoes hepatic me-
`tabolism with limited unchanged drug
`excreted in the urine or stool.
`Cunningham et al. (13) evaluated
`dexmedetomidine pharmacokinetics fol-
`lowing administration (0.6 ␮g/kg infused
`over 10 mins) in five adults with severe
`
`hepatic failure and compared the results
`with five age-matched controls with nor-
`mal hepatic function. When compared
`with age-matched controls with normal
`hepatic function, there was an increased
`volume of distribution at steady state (3.2
`vs. 2.2 L/kg, p ⬍ .05), an increased elim-
`ination half-life (7.5 vs. 2.6 hrs, p ⬍ .05),
`and a decreased clearance (0.32 vs. 0.64
`L/hr/kg; p ⬍ .05) in patients with hepatic
`dysfunction. In a subsequent study in six
`adult patients with severe renal disease
`(24-hr creatinine clearance ⱕ30 mL/min)
`who were not receiving dialysis, there
`was no statistically significant difference
`between renal disease and control pa-
`
`116
`
`Pediatr Crit Care Med 2007 Vol. 8, No. 2
`
`Petition for Inter Partes Review of US 8,455,527
`Amneal Pharmaceuticals LLC – Exhibit 1061 – Page 116
`
`

`

`tients in the volume of distribution at
`steady state (1.81 ⫾ 0.55 vs. 1.54 ⫾ 0.08
`L/kg) or the elimination clearance
`(12.5 ⫾ 4.6 vs. 8.9 ⫾ 0.7 mL/min/kg)
`(14). However, the elimination half-life
`was decreased with renal disease (113.4 ⫾
`11.3 mins vs. 136.5 ⫾ 13.0 mins, p ⬍
`.05). Despite the shorter elimination half-
`life, there was prolonged sedation in pa-
`tients with renal disease. The 1-hr postin-
`fusion visual analog score of sedation
`(scale of 0 to 100) was 49.2 ⫾ 25.4 in
`patients with renal disease compared
`with 26.2 ⫾ 18.3 in patients with normal
`function (p ⬍ .05). The authors
`renal
`speculated that the increased sedation
`with renal failure resulted from decreased
`protein binding and an increased free
`fraction of the drug. Venn et al. (15) eval-
`uated the impact of acute surgical inter-
`vention and critical illness on dexmedeto-
`midine pharmacokinetics in ten adult
`patients following complex abdominal or
`pelvic surgical procedures. Dexmedeto-
`midine administration included a loading
`dose of 0.4 ␮g/kg over 10 mins followed
`by an infusion of 0.7 ␮g/kg/hr. When
`compared with data from healthy volun-
`teers, there was no difference in half-life,
`volume of distribution, or clearance.
`Data regarding dexmedetomidine
`pharmacokinetics in the pediatric popu-
`lation have been presented in one recent
`manuscript and two abstracts (16 –18).
`Petroz et al. (16) randomized 36 children,
`ranging in age from 2 to 12 yrs, to receive
`dexmedetomidine infused for 10 mins at
`2, 4, or 6 ␮g/kg/hr (0.33, 0.6, and 1 ␮g/
`kg). Using a two-compartment model,
`they reported that the pharmacokinetics
`of dexmedetomidine in children are sim-
`ilar to adults with no dose-dependent ki-
`netics, protein binding of 92.6%, weight-
`adjusted total body clearance of 13 mL/
`kg/min, a volume of distribution of the
`peripheral compartment of 1.0 L/kg, and
`a terminal elimination half-life of 1.8 hrs.
`Rodarte et al. (17) administered a contin-
`uous infusion in a dose ranging from 0.2
`to 0.7 ␮g/kg/hr for 8 –24 hrs to ten chil-
`dren (0.3–7.9 yrs of age) following cardiac
`procedures (n ⫽ 9) or craniofacial proce-
`dures (n ⫽ 1). Using a two-compartment
`model, they reported a volume of distri-
`bution of 1.53 ⫾ 0.37 L/kg, a clearance of
`0.57 ⫾ 0.14 L/kg/hr (approximately 9.5
`mL/kg/min), and a terminal elimination
`half-life of 2.65 ⫾ 0.88 hrs. They com-
`mented that their data demonstrated that
`the pharmacokinetics of dexmedetomi-
`dine in children were predictable and
`consistent with results reported in adults.
`
`Pediatr Crit Care Med 2007 Vol. 8, No. 2
`
`The final pharmacokinetic study in chil-
`dren included infants, ranging in age
`from 1 to 24 months, following surgery
`for congenital heart disease (18). The au-
`thors reported a median clearance of 27.2
`mL/kg/min, peripheral volume of distri-
`bution of 2.5 L/kg, and terminal elimina-
`tion half-life of 83 mins. They concluded
`that infants appear to clear dexmedeto-
`midine more quickly than adults or older
`children.
`
`END-ORGAN EFFECTS OF
`DEXMEDETOMIDINE
`
`Cardiovascular and
`Hemodynamic Effects
`
`Heart Rate, Blood Pressure, Cardiac
`Output, and Myocardial Contractility. Hy-
`potension and bradycardia have been re-
`ported in adult patients, especially in the
`presence of comorbid cardiac disease,
`when administered with other medica-
`tions that possess negative chronotropic
`effects or following large or rapid bolus
`doses. In healthy adult volunteers, there
`is a biphasic effect following dexmedeto-
`midine with an initial increase in systolic
`blood pressure (sBP) and a reflex decrease
`in HR followed by a stabilization of sBP
`and HR at values below the baseline (19).
`Stimulation of peripheral postsynaptic
`␣2B-adrenergic receptors results in vaso-
`constriction and the initial increase in
`sBP, whereas the eventual decrease in BP
`and HR results from central presynaptic
`␣2A-adrenergic receptor stimulated sym-
`patholysis.
`In healthy, adult volunteers, dexme-
`detomidine doses of 0.25, 0.5, 1.0 and 2.0
`␮g/kg administered over 2 mins resulted
`in a decrease from baseline of the mean
`arterial pressure (MAP) at 60 mins of
`14%, 16%, 23%, and 27% (19). Following
`a dose of 1 ␮g/kg, cardiac output, mea-
`sured by thoracic bioimpedance, was
`81 ⫾ 13% of baseline at 1 min, 88 ⫾ 14%
`of baseline at 10 mins, and 91 ⫾ 11% of
`baseline at 60 mins. With a dose of 2
`␮g/kg, cardiac output was 58 ⫾ 32% of
`baseline at 1 min, 76 ⫾ 33% of baseline
`at 10 mins, and 85 ⫾ 28% of baseline at
`60 mins.
`for adverse hemody-
`The potential
`namic effects with dexmedetomidine in
`patients with comorbid features is illus-
`trated in an adult ICU population of 98
`cardiac and general surgery patients who
`received dexmedetomidine for sedation
`during mechanical ventilation (11).
`
`Dexmedetomidine was dosed as a bolus
`dose of 1 ␮g/kg over 10 mins followed by
`an infusion of 0.2– 0.7 ␮g/kg/hr. Hypo-
`tension (MAP ⱕ60 mm Hg or a ⱖ30%
`decrease from baseline) occurred in 18 of
`66 patients. Eleven of the episodes oc-
`curred during the bolus. Hypertension
`was noted in six of the 66 patients during
`the loading dose. Although no morbidity
`or mortality was noted, the infusion was
`temporarily (n ⫽ 3) or permanently (n ⫽
`3) discontinued, and treatment with at-
`ropine (n ⫽ 2) or temporary cardiac pac-
`ing (n ⫽ 4) was necessary.
`Bradycardia and sinus arrest have
`been reported with dexmedetomidine
`(20, 21). In a study combining dexme-
`detomidine with propofol to induce anes-
`thesia, two of the first four patients had
`brief and self-limited sinus arrest after
`laryngoscopy (20). Dexmedetomidine was
`administered as a bolus dose of 1 ␮g/kg
`over 15 mins followed by an infusion of
`0.4 ␮g/kg/hr resulting in the administra-
`tion of an average dose of 1.47 ␮g/kg
`before anesthetic induction with propo-
`fol. The protocol was amended (decrease
`of the dexmedetomidine dose to 0.7
`␮g/kg over 15 mins followed by an infu-
`sion of 0.27 ␮g/kg/hr), and no subsequent
`problems were noted.
`We reported bradycardia in a 5-wk-old
`infant with trisomy 21 who was receiving
`dexmedetomidine for sedation during
`mechanical ventilation (22). Concomi-
`tant medications included digoxin for the
`treatment of chronic congestive heart
`failure due to an unrepaired atrioventric-
`ular canal defect. Twelve hours after the
`initiation of the dexmedetomidine infu-
`sion, the infant’s HR decreased to 40 –50
`beats/min with a stable BP. The dexme-
`detomidine infusion was discontinued
`without other therapy, and the HR re-
`turned to baseline within 60 mins.
`In a study of 192 patients with Amer-
`ican Society of Anesthesiologists ratings
`of 1 or 2, randomized to receive either
`intramuscular dexmedetomidine and in-
`travenous saline, intramuscular dexme-
`detomidine and intravenous fentanyl, or
`intramuscular midazolam and intrave-
`nous fentanyl, followed by maintenance
`anesthesia (70% nitrous oxide in 30%
`oxygen, fentanyl, and either enflurane or
`isoflurane),
`intraoperative bradycardia
`and hypotension were significantly more
`common in the patients who received
`dexmedetomidine compared with those
`receiving midazolam (23). In one patient,
`bradycardia (HR 35 beats/min) required
`pharmacologic therapy. Khan et al. (24),
`
`117
`
`Petition for Inter Partes Review of US 8,455,527
`Amneal Pharmaceuticals LLC – Exhibit 1061 – Page 117
`
`

`

`in a study of nine male volunteers assess-
`ing the effects of low (0.3 ng/mL) and
`high (0.6 ng/mL) dexmedetomidine
`plasma concentrations on isoflurane re-
`quirements, reported five hypotensive
`events in the low concentration group
`and seven in the concentration group.
`Interventions including crystalloid, crys-
`talloid and methoxamine, or atropine
`were necessary in five patients. The ma-
`jority of the hemodynamic events (75%)
`occurred at an end-tidal
`isoflurane of
`ⱖ1%.
`In a cohort of 80 children, ranging in
`age from 1 to 12 yrs, no clinically signif-
`icant hypotension or bradycardia oc-
`curred with the intraoperative adminis-
`tration of dexmedetomidine (0.5 ␮g/kg)
`during anesthesia at 1 minimum alveolar
`concentration with either desflurane or
`sevoflurane (25). However, there was a
`greater decrease in HR in patients anes-
`thetized with sevoflurane compared with
`those receiving desflurane (104 ⫾ 16 vs.
`120 ⫾ 17 beats/min, p ⬍ .01).
`Lowering of HR and thereby myocar-
`dial oxygen consumption may provide
`beneficial effects in patients with coro-
`nary artery disease. Talke et al. (26) ran-
`domized 24 adult patients undergoing
`vascular surgery to placebo or one of
`three plasma concentrations of dexme-
`detomidine: 0.15 ng/mL (low dose), 0.3
`ng/mL (medium dose), or 0.45 ng/mL
`(high dose). Dexmedetomidine was
`started 1 hr before anesthetic induction
`and continued for 48 hrs. Although there
`was an increased intraoperative need for
`atropine and/or phenylephrine with
`dexmedetomidine, no such difference was
`noted postoperatively. In the placebo
`group, there was an increased incidence of
`tachycardia (23 mins/hr) when compared
`with the low-dose (9 mins/hr, p ⫽ .006),
`medium-dose (0.5 mins/hr, p ⫽ .004),
`and high-dose (2.3 mins/hr, p ⫽ .004)
`dexmedetomidine groups. In an anec-
`dotal report, the negative chronotropic
`effect of dexmedetomidine was used as a
`therapeutic maneuver during off-pump
`coronary artery bypass surgery when
`tachycardia was unresponsive to ␤-adren-
`ergic blockade (27).
`The potential for significant negative
`chronotropic effects appears to be greater
`when dexmedetomidine is administered
`with medications that have negative
`chronotropic effects (propofol, succinyl-
`choline, digoxin, pyridostigmine) or dur-
`ing vagotonic procedures (laryngoscopy)
`(20 –22). Animal studies have not demon-
`strated direct effects on myocardial con-
`
`118
`
`tractility or intracellular calcium regula-
`tion (28). When studied in an isolated
`right ventricular papillary muscle prepa-
`ration, dexmedetomidine had no effect on
`the amplitude and time variables of iso-
`metric, isotonic, or zero-loaded-clamped
`twitches and intracellular calcium cur-
`rents (28).
`Sympathetic Nervous System and En-
`dogenous Catecholamine Release. Bio-
`chemical data from a cohort of eight
`adult postoperative patients demonstrate
`the sympatholytic effects of dexmedeto-
`midine (29). Following a 60-min dexme-
`detomidine infusion administered by a
`computer-controlled infusion protocol
`to achieve a plasma concentration of
`600 pg/mL, the plasma norepinephrine
`concentration decreased from 2.1 ⫾ 0.8
`to 0.7 ⫾ 0.3 nmol/L, the plasma epi-
`nephrine concentration decreased from
`0.7 ⫾ 0.5 to 0.2 ⫾ 0.2 nmol/L, HR
`decreased from 76 ⫾ 15 to 64 ⫾ 11
`beats/min, and sBP decreased from 158
`⫾ 23 to 140 ⫾ 23 mm Hg. The same
`investigators evaluated changes in
`plasma and urinary catecholamines in
`41 adult patients undergoing vascular
`surgery (30). Dexmedetomidine was
`started intraoperatively and continued
`for the first 48 postoperative hours.
`When compared with patients receiving
`dexmedetomidine, plasma norepineph-
`rine concentrations were two to three
`times higher at the time of tracheal
`extubation and at 60 mins after arrival
`in the postanesthesia care unit than in
`the control group. Urinary normeta-
`nephrine levels increased significantly
`in the placebo group, whereas no
`change was noted in patients receiving
`dexmedetomidine.
`A similar sympatholytic effect has
`been demonstrated following the intraop-
`erative administration of dexmedetomi-
`dine to pediatric patients undergoing car-
`diopulmonary bypass and surgery for
`congenital heart disease (31). Muktar et
`al. (31) randomized 30 infants and chil-
`dren to placebo or dexmedetomidine (bo-
`lus of 10 ␮g/kg over 10 mins followed by
`an infusion of 0.5 ␮g/kg/hr), which was
`administered after anesthetic induction
`and placement of arterial and venous can-
`nulae. Although plasma cortisol, norepi-
`nephrine, epinephrine, and glucose con-
`centrations
`increased in both the
`dexmedetomidine and the placebo groups
`after sternotomy and following cardiopul-
`monary bypass, the increase was signifi-
`cantly less in patients receiving dexme-
`detomidine. Additionally, when weaning
`
`less so-
`from cardiopulmonary bypass,
`dium nitroprusside was required in pa-
`tients receiving dexmedetomidine (0.3 ⫾
`0.36 vs. 1.3 ⫾ 0.68 ␮g/kg/min, p ⬍ .05).
`No adverse effects were noted.
`In specific clinical scenarios such as
`hemorrhage, hypovolemia, or conges-
`tive heart failure, there is the potential
`for dexmedetomidine’s sympatholytic
`effect to be detrimental by offsetting
`the protective function of the sympa-
`thetic nervous system. In an animal
`model, Blake et al. (32) evaluated the
`effects of dexmedetomidine on the BP
`response during incremental decreases
`in intravascular blood volume induced
`by a gradual
`inflation of an inferior
`vena cava cuff. In control animals, the
`gradual reduction of intravascular vol-
`ume resulted in a progressive increase
`in HR with peripheral vasoconstriction
`to maintain MAP until cardiac index
`was approximately 40% of baseline, at
`which time there was failure of vaso-
`constriction and a decrease in MAP.
`Dexmedetomidine, administered intra-
`venously or directly into the fourth
`ventricle of the central nervous system,
`resulted in a decrease of HR and MAP
`from baseline and an earlier decompen-
`sation during inflation of the inferior
`vena cava cuff. Similar findings were
`reported in rabbits treated with doxo-
`rubicin to induce a chronic congestive
`heart failure and then subjected a re-
`duction of intravascular volume by in-
`flation of an inferior vena cava cuff (33).
`Myocardial Oxygen Consumption and
`Perioperative Ischemia. Clinical studies
`in adults have shown that the periopera-
`tive administrative of ␣2-adrenergic ago-
`nists may modify the incidence of adverse
`cardiovascular events including myocar-
`dial
`ischemia (34, 35). In an animal
`model of coronary artery stenosis,
`dexmedetomidine reduced blood flow in
`the nonischemic myocardium and in the
`ischemic epicardial layer; however, there
`was no effect on blood flow in the isch-
`emic mid-myocardial and subendocardial
`layers, thereby increasing the ischemic-
`nonischemic blood flow ratio (36). Myo-
`cardial oxygen demand also decreased
`with dexmedetomidine, thereby further re-
`ducing the ischemic myocardium’s oxygen
`deficiency
`Similar findings were reported by
`Willigers et al. (37) in their animal model
`using graded coronary stenosis to pro-
`duce lactate release from the poststenotic
`myocardium. Lactate production oc-
`curred in zero of eight dogs receiving
`
`Pediatr Crit Care Med 2007 Vol. 8, No. 2
`
`Petition for Inter Partes Review of US 8,455,527
`Amneal Pharmaceuticals LLC – Exhibit 1061 – Page 118
`
`

`

`dexmedetomidine compared with four of
`seven in the control group (p ⫽ .03).
`With dexmedetomidine,
`lactate release
`was 46% less during emergence from an-
`esthesia, and the endocardial/epicardial
`blood flow ratio increased by 35% com-
`pared with the control group. Decreased
`levels of plasma epinephrine (158 vs.
`1909 pg/mL) and norepinephrine (126 vs.
`577 pg/mL) and decreased HR (123 ⫾ 6
`vs. 160 ⫾ 10 beats/min) were noted. The
`authors postulated that this may account
`for the anti-ischemic effect of dexmedeto-
`midine.
`Additional potentially protective ef-
`fects of dexmedetomidine on myocar-
`dial performance include preservation
`of myocardial dysfunction following
`ischemia and prevention of catechol-
`amine-induced arrhythmogenesis (38,
`39). Hypoxia followed by reoxygenation
`exposes the myocardium to an oxidative
`stress, resulting in tissue injury/death
`and myocardial dysfunction. In rats ex-
`posed to 60 mins of hypoxia, dexme-
`detomidine administered before but not
`after hypoxia significantly improved left
`ventricular-developed pressure after
`reoxygenation (38). The effect was
`blocked by yohimbine, an ␣2-adrenergic
`antagonist. In a separate study, dexme-
`detomidine increased the dysrhythmo-
`genic dose of epinephrine in halothane-
`anesthetized dogs (mean dose of 3 ␮g/kg/
`min in control animals vs. 6 ␮g/kg/min in
`animals receiving dexmedetomidine) (39).
`Pulmonary Vascular Resistance.
`There is limited information regarding
`dexmedetomidine’s effects on the pulmo-
`nary vasculature and pulmonary vascular
`resistance (PVR). In six instrumented
`sheep, dexmedetomidine (2 ␮g/kg over 1
`min) transiently increased mean pulmo-
`nary artery pressure (MPAP) and PVR (40).
`PVR increased from a baseline of 81 ⫾ 16
`dynes·sec·cm⫺5 to a maximum of 141 ⫾
`27 dynes·sec·cm⫺5, whereas MPAP in-
`creased from 15 ⫾ 1 to 18⫾ 0 mm Hg.
`MAP also increased (86 ⫾ 2 to 93⫾ 6
`mm Hg), as did systemic vascular resis-
`tance (1416 ⫾ 83 to 1889 ⫾ 64
`dynes·sec·cm⫺5). There was no change in
`pulmonary artery occlusion pressure.
`Similar transient pulmonary hemody-
`namic changes have been reported in
`healthy human volunteers with graded
`dexmedetomidine infusions to a plasma
`concentration of 1.9 ng/mL (19). Given
`the potential impact of these findings,
`especially in patients with elevated MPAP
`
`Pediatr Crit Care Med 2007 Vol. 8, No. 2
`
`or PVR, future studies are needed to de-
`fine these effects.
`
`Respiratory Effects
`
`Ventilation. The ventilatory effects of
`increasing doses of dexmedetomidine
`(0.25, 0.5, 1, and 2 ␮g/kg over 2 mins)
`have been evaluated in healthy adult vol-
`unteers by measurement of oxygen satu-
`ration, PaCO2, CO2 response curves with
`CO2 rebreathing, and respiratory induc-
`tance plethysmography (10, 41). With
`doses of 1 or 2 ␮g/kg, PaCO2 increased
`significantly with a maximum effect
`noted 10 mins following the dose. The
`mean PaCO2 increase from baseline was
`5.0 and 4.2 mm Hg with the 1.0 and 2.0
`␮g/kg doses, respectively. The effect per-
`sisted for 60 mins following 1 ␮g/kg and
`for 105 mins following 2 ␮g/kg. Follow-
`ing 2.0 ␮g/kg, minute ventilation de-
`creased from 8.7 ⫾ 0.7 to 6.3 ⫾ 1.5 L/min
`(p ⬍ .05). The decrease resulted predom-
`inantly from a decreased tidal volume
`with less effect on respiratory rate. Sig-
`nificant changes were also noted using
`CO2 response curves, as minute ventila-
`tion at an end-tidal CO2 of 55 mm Hg was
`depressed following the both the 1- and
`2-␮g/kg doses. The authors also noted
`short episodes of apnea and irregular
`breathing in some subjects, which oc-
`curred more commonly with the two
`highest doses (seven of ten patients with
`2 ␮g/kg and five of six patients with 1
`␮g/kg vs. one of six with either 0.5 ␮g/kg
`or 0.25 ␮g/kg). Respiratory inductance
`plethysmography was used to demon-
`strate that these problems were obstruc-
`tive and not central. Although oxygen
`saturation decreased with the obstructive
`episodes, the average room air oxygen
`saturation remained ⬎95% following all
`doses of dexmedetomidine. The oxygen
`saturation decrease was greatest at 10
`mins following 1 ␮g/kg (decrease from
`98.5 ⫾ 0.7% to 96.2 ⫾ 1.3%) and at 60
`mins following 2 ␮g/kg (decrease from
`98.3 ⫾ 0.8% to 95.4 ⫾ 1.2%). Similar
`respiratory effects have been demon-
`strated in experimental animals although
`a paradoxic effect has been noted with
`more of an effect on ventilation with 1 vs.
`10 ␮g/kg in one study and 10 or 30 ␮g/kg
`vs. 50 ␮g/kg in another (42– 44).
`Conflicting results were reported
`when comparing the respiratory effects of
`dexmedetomidine with remifentanil
`in
`six healthy adult volunteers (45). When
`compared with baseline, a remifentanil
`infusion to achieve a stepwise plasma
`
`concentration of 1, 2, 3, and 4 ng/mL
`resulted in respiratory depression mani-
`fested as a decrease in respiratory rate
`and minute ventilation, increased PaCO2,
`blunting of the CO2 response curve, and
`apnea with oxygen desaturation. During
`stepwise dexmedetomidine infusions to
`achieve plasma concentrations of 0.6, 1.2,
`1.8, and 2.4 ng/mL, there was an increase
`in respiratory rate, a decrease in the hy-
`popnea/apnea index, and no change in
`the end-tidal CO2 when compared with
`baseline values. With dexmedetomidine,
`some patients demonstrated a periodic in-
`crease in minute ventilation during CO2
`response curves (hypercapnic arousal) that
`correlated with changes in the Bispectral
`Index. The authors noted that similar
`changes occur during natural sleep and
`that these findings may result from
`dexmedetomidine’s mechanism of action
`in the locus ceruleus and its convergence
`on the natural sleep pathway. The au-
`thors concluded that dexmedetomidine
`stands apart from other sedatives in that
`it appears to be clinically safe from a
`respiratory point of view even in doses
`high enough to cause unresponsiveness.
`Similar findings were reported from an
`evaluation of the respiratory effects of
`dexmedetomidine (10 and 30 ␮g/kg) and
`alfentanil in an animal model (rats) (46).
`Neither dose of dexmedetomidine had an
`effect on PaO2, PaCO2, or pH, whereas the
`administration of alfentanil resulted in a
`decrease in pH and PaO2 and an increase in
`PaCO2. Dexmedetomidine had no additional
`effect when administered after alfentanil,
`and in fact, dexmedetomidine in a dose of
`30 ␮g/kg decreased the acidosis and hyper-
`capnia that occurred following alfentanil.
`Despite these findings, monitoring of respi-
`ratory function during the administration
`of dexmedetomidine in high-risk patients
`or those receiving other agents that may
`depress respiratory function appears war-
`ranted given the recent report of central
`apnea after a general anesthetic that in-
`cluded dexmedetomidine (47).
`Airway Reactivity. In mongrel dogs,
`the intravenous but not the inhalational
`administration of dexmedetomidine has
`been shown to prevent histamine-in-
`duced bronchoconstriction (48). Bron-
`choconstriction was provoked with aero-
`solized histamine, and its effect on airway
`caliber was evaluated using high-resolu-
`tion computed tomography with an eval-
`uation of airway cross-sectional area.
`Aerosolized histamine constricted the
`airways to 66 ⫾ 27% of baseline com-
`pared with 87 ⫾ 30.4% of baseline when
`
`119
`
`Petition for Inter Partes Review of US 8,455,527
`Amneal Pharmaceuticals LLC – Exhibit 1061 – Page 119
`
`

`

`the animals were pretreated with intrave-
`nous dexmedetomidine.
`
`Central Nervous System Effects
`
`Sedation. Clinical studies in humans
`and experimental trials in both humans
`and animals have demonstrated the sed-
`ative effects of dexmedetomidine (9, 10,
`41, 49, 50). In ten healthy adult male
`volunteers, sequential 40-min infusions
`of dexmedetomidine were administered
`to achieve plasma concentrations of 0.5,
`0.8, 1.2, 2.0, 3.2, 5.0, and 8.0 ng/mL (45).
`The visual analog sedation score (0 ⫽
`very alert and 100 ⫽ very sedated) in-
`creased to 36 ⫾ 27 and 62 ⫾ 18 from a
`baseline of 0 with the first two targeted
`infusion levels (0.5 and 0.8 ng/mL). The
`two volunteers who received the highest
`incremental dose (calculated to achieve a
`plasma concentration of 8.0 ng/mL) were
`not arousable even with vigorous shak-
`ing. Picture recall and recognition were
`preserved during the lowest incremental
`infusion (0.5 ng/mL) but were 0% (0 of
`10) and 20% (2 of 10), respectively, with
`the second and third infusion levels (0.8
`and 1.2 ng/mL).
`Dexmedetomidine’s sedative response
`has been shown to have properties that
`parallel natural sleep (49, 50). Using
`functional magnetic resonance imaging
`(MRI), the blood oxygen level dependent
`signal, a correlate of local brain activity,
`changes with dexmedetomidine-induced
`sedation in a similar fashion to that seen
`during natural sleep (50). This is different
`from the pattern that occurs following
`the administration of midazolam. Using
`immunohistochemistry and in situ hy-
`bridization, dexmedetomidine has also
`been shown to induce a qualitatively sim-
`ilar pattern of c-fos expression in sleep-
`promoting brain nuclei of rats as that
`seen during nonrapid eye movement
`sleep (a decrease in the locus ceruleus
`and tuberomammillary nucleus and an
`increase in the ventrolateral nucleus)
`(49). These effects were attenuated by ati-
`pamezole, an ␣2-adrenergic antagonist,
`and did not occur in rats that lacked
`␣2-adrenergic receptors. These findings
`suggest a clinical advantage of dexmedeto-
`midine with its pattern of sedation parallel-
`ing natural sleep when compared with
`other agents (barbiturates, benzodiaz-
`epines, and propofol) commonly used for
`ICU sedation. These agents disrupt the nor-
`mal electroencephalographic patterns of
`sleep, and these effects may be responsible
`for the delirium seen in the ICU setting.
`
`120
`
`Given that delirium has been shown to be
`an independent risk factor of mortality in
`the adult ICU setting, avoidance of the dis-
`ruption of the natural sleep cycle

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