throbber
Cardiac Intensive Care
`
`Dexmedetomidine use in a pediatric cardiac intensive care unit:
`Can we use it in infants after cardiac surgery?
`
`Constantinos Chrysostomou, MD; Joan Sanchez De Toledo, MD; Tracy Avolio, CCRN; Maria V. Motoa, MD;
`Donald Berry, BS, RPh; Victor O. Morell, MD; Richard Orr, MD; Ricardo Munoz, MD
`
`Objective: To assess clinical response of dexmedetomidine
`alone or in combination with conventional sedatives/analgesics
`after cardiac surgery.
`Design: Retrospective study.
`Setting: Pediatric cardiac intensive care unit.
`Patients: Infants and neonates after cardiac surgery.
`Measurements and Main Results: We identified 80 patients
`including 14 neonates, at mean age and weight of 4.1 ⴞ 3.1
`months and 5.5 ⴞ 2 kg, respectively, who received dexmedeto-
`midine for 25 ⴞ 13 hours at an average dose of 0.66 ⴞ 0.26
`␮g䡠kgⴚ1䡠hrⴚ1. Overall normal sleep to moderate sedation was
`documented 94% of the time and no pain to mild pain for 90%.
`Systolic blood pressure (SBP) decreased from 89 ⴞ 15 mm Hg to
`85 ⴞ 11 mm Hg (p ⴝ .05), heart rate (HR) from 149 ⴞ 22 bpm to
`129 ⴞ 16 bpm (p < .001), and respiratory rate (RR) remained
`unchanged. When baseline arterial blood gases were compared
`with the most abnormal values, pH decreased from 7.4 ⴞ 0.07 to
`7.37 ⴞ 0.05 (p ⴝ .006), PO2 from 91 ⴞ 67 mm Hg to 66 ⴞ 29
`mm Hg (p ⴝ .005), and CO2 increased from 45 ⴞ 8 mm Hg to 50ⴞ
`12 mm Hg (p ⴝ .001). At the beginning of the study, 37 patients
`(46%) were mechanically ventilated; and at 48 hours, 13 patients
`
`intubated and five patients failed extubation.
`(16%) were still
`Three groups of patients were identified: A, dexmedetomidine
`only (n ⴝ 20); B, dexmedetomidine with sedatives/analgesics
`(n ⴝ 38); and C, dexmedetomidine with both sedatives/analgesics
`and fentanyl infusion (n ⴝ 22). The doses of dexmedetomidine
`and rescue sedatives/analgesics were not significantly different
`among the three groups but duration of dexmedetomidine was
`longer in group C vs. A (p ⴝ .03) and C vs. B (p ⴝ .002). Pain,
`sedation, SBP, RR, and arterial blood gases were similar. HR was
`higher in group C vs. B (p ⴝ .01). Comparison between neonates
`and infants showed that infants required higher dexmedetomidine
`doses, 0.69 ⴞ 25 ␮g䡠kgⴚ1䡠hrⴚ1, and vs. 0.47 ⴞ 21 ␮g䡠kgⴚ1䡠hrⴚ1
`(p ⴝ .003) and had lower HR (p ⴝ .01), and RR (p ⴝ .009), and
`higher SBP (p < .001).
`Conclusions: Dexmedetomidine use in infants and neonates
`after cardiac surgery was well tolerated in both intubated and
`nonintubated patients. It provides an adequate level of sedation/
`analgesia either alone or in combination with low-dose conven-
`tional agents. (Pediatr Crit Care Med 2009; 10:654 – 660)
`KEY WORDS: sedation; analgesia; infants; dexmedetomidine; car-
`diac surgery; intensive care unit; neonates
`
`P roviding sedation and analge-
`
`sia in children after cardiac
`surgery can be challenging. Al-
`though our knowledge about
`sedative agents and their cardiorespira-
`tory interactions has improved during
`the last two decades, delivering optimal
`sedation in the postoperative period re-
`
`From the Department of Pediatrics and Critical
`Care Medicine, Division of Cardiac Intensive Care (CC,
`JSDT, TA, RM, DO, MVM, DB) and Department of
`Cardiothoracic Surgery (VOM), Children’s Hospital of
`Pittsburgh of the University of Pittsburgh Medical Cen-
`ter, Pittsburgh, PA.
`Supported, partially, by Hospira, Inc. Lake Forest, IL.
`This study was performed at Cardiac Intensive
`Care Unit, Children’s Hospital of Pittsburgh of
`the
`University of Pittsburgh Medical Center.
`The authors have not disclosed any potential con-
`flicts of interest.
`For information regarding this article, E-mail:
`chrycx@chp.edu
`Copyright © 2009 by the Society of Critical Care
`Medicine and the World Federation of Pediatric Inten-
`sive and Critical Care Societies
`DOI: 10.1097/PCC.0b013e3181a00b7a
`
`654
`
`mains complex. Some of the factors that
`add to this complexity are the presence of
`an unpredictable and potentially labile
`physiology after cardiopulmonary bypass
`and the inability to accurately assess an
`infant’s level of sedation.
`In our institution, a successful effort
`for fast track and early extubation has
`been in place for the last 5 yrs. Nonethe-
`less, a significant proportion of patients
`remain, who could further benefit from
`newer sedative agents with a better safety
`profile and less respiratory depression.
`This is important for infants with both
`univentricular and biventricular physiol-
`ogy and particularly the ones which are
`extubated or near extubation. Dexme-
`detomidine is a highly specific alpha-2
`adrenergic receptor agonist with seda-
`tive, analgesic, and anxiolytic properties
`(1). It does not appear to significantly
`depress respiratory drive, thus interfer-
`ence with weaning from mechanical ven-
`tilation is less likely. In fact, it has been
`used both as a bridge to extubation as
`
`well as in nonintubated patients (2, 3). In
`our previous study, which was mainly
`focused on older children after cardiotho-
`racic surgery, dexmedetomidine was
`found to be well tolerated and provided
`targeted sedation and analgesia for 93%
`and 83% of the time, respectively (2). We
`describe our experience with the use of
`dexmedetomidine in a much younger and
`rather more difficult population: Infants,
`and neonates after congenital cardiac
`surgery.
`
`MATERIALS AND METHODS
`
`This retrospective case series study was
`approved by the Institutional Review Board of
`the University of Pittsburgh Medical Center/
`Children’s Hospital of Pittsburgh. To follow
`our institution’s policy, parental
`informed
`consent for investigational use of a drug was
`obtained for all patients younger than 1 year.
`Infants and neonates who were admitted to
`the cardiac intensive care unit (CICU) from
`January 2004 to May 2007 and had received
`dexmedetomidine were included.
`
`Pediatr Crit Care Med 2009 Vol. 10, No. 6
`
`Petition for Inter Partes Review of US 8,338,470
`Amneal Pharmaceuticals LLC – Exhibit 1045 – Page 654
`
`

`
`Dexmedetomidine was started as a con-
`tinuous infusion, at a dose of 0.1–1.25
`␮g䡠kg⫺1䡠hr⫺1. If it was considered necessary, a
`1 ␮g/kg bolus at a rate of 0.1 ␮g䡠kg⫺1䡠min⫺1
`was given. According to the clinical practice in
`our CICU, the decision to give a bolus and the
`exact initial infusion dose was based on the
`physician’s judgment and the level of sedation
`that the patient had before the initiation of the
`infusion, i.e., the degree of residual intraoper-
`ative anesthesia. Twenty minutes after the on-
`set of the infusion, if sedation/analgesia were
`considered inadequate by the bedside nurse
`and by the physician on duty, dexmedetomi-
`dine infusion was increased by 0.1– 0.3
`␮g䡠kg⫺1䡠hr⫺1. If sedation/analgesia was still
`inadequate 20 minutes after the change of the
`infusion dose, a rescue agent was adminis-
`tered if necessary, and dexmedetomidine infu-
`sion was further increased to a maximum of
`1.5 ␮g䡠kg⫺1䡠hr⫺1. The maximum infusion
`dose of 1.5 ␮g䡠kg⫺1䡠hr⫺1 was a consensus de-
`cision among the intensivists and pharmacists
`involved with the care of CICU patients. Given
`the lack of an established sedation protocol
`and the diversity of our patient population, the
`rescue agents were multiple and included fen-
`tanyl, morphine, midazolam, lorazepam, ket-
`amine, and chloral hydrate.
`Heart rate (HR), systolic blood pressure
`(SBP), respiratory rate (RR), and sedation and
`analgesia scores were recorded by the nursing
`staff at baseline, i.e., measurement within 1
`hour before starting dexmedetomidine, and
`every hour thereafter. Arterial blood gas (ABG)
`results also were recorded every 1– 4 hrs as
`clinically indicated. A hypertensive or hypo-
`tensive episode was defined as a 30% change
`from baseline and/or if the SBP was below or
`
`Table 1. Cardiac diagnosis/procedures performed
`
`Tetralogy of Fallot
`Coarctation of the aorta
`VSD with or without ASD
`Atrioventricular septal defect
`Glenn
`Hypoplastic or interrupted aortic
`arch and VSD
`Norwood stage I (HLHS)
`Total anomalous pulmonary venous
`return
`Pulmonary valvotomy and VSD
`Blalock-Taussig shunt
`Transposition of the great arteries
`Aortopulmonary window
`Right ventricle to pulmonary artery
`conduit
`Left pulmonary artery augmentation
`Othera
`
`n (%)
`
`19 (24)
`14 (17.5)
`12 (15)
`9 (11)
`9 (11)
`3 (4)
`
`2 (2.5)
`2 (2.5)
`
`2 (2.5)
`2 (2.5)
`1
`1
`1
`
`1
`2
`
`ASD, atrial septal defect; HLHS, hypoplastic left
`heart syndrome; VSD, ventricular septal defect.
`aOne patient s/p cardiac catheterization and
`one with severe pulmonary hypertension.
`
`Pediatr Crit Care Med 2009 Vol. 10, No. 6
`
`above the 5th to 95th percentile for age. A
`bradycardic episode was defined as a 30%
`change from baseline and/or if the HR was
`below the fifth percentile for age. Low RR rate
`was defined as a RR below the fifth percentile
`for age. Sedation was assessed using a pediat-
`ric intensive care unit sedation scale ranging
`from 0 to 3 (0, none, alert, or normal sleep,
`easy to arouse; 1, mild sedation, occasionally
`drowsy, easy to arouse; 2, moderate sedation,
`frequently drowsy but easy to arouse; 3, severe
`sedation, somnolent, difficult to arouse). An-
`algesia was assessed with two 0 –10 pain score
`scales: The FLACC (face, legs, activity, cry, and
`consolability) for patients older than 2 months
`old, and the CRIES (cries, requires oxygen for
`saturation less than 95%, increased vital signs,
`expression, sleepless) for patients 0 – 6 months
`old (4, 5). A pain score of 0 was considered
`pain free, 1–3 mild pain, 4 –7 moderate pain,
`and 8 –10 severe pain. The targeted level of
`sedation and analgesia was 0 –2 and 0 –3, re-
`spectively. Data were collected for as long as
`the dexmedetomidine infusion was being ad-
`ministered to a maximum of 48 hours.
`Absolute exclusion criteria for the use of
`dexmedetomidine included decompensated
`heart failure, acute hemodynamic instability,
`septic shock, and ventricular arrhythmias.
`Relative exclusion criteria included a high
`RACHS-1 (risk adjustment for congenital
`heart surgery) score, 5 and 6 (6).
`Patients were divided into three groups: A,
`dexmedetomidine only (n ⫽ 20); B, dexme-
`detomidine with sedatives/analgesics (n ⫽ 38);
`and C, dexmedetomidine with both sedatives/
`infusion (n ⫽ 22).
`analgesics and fentanyl
`Three major comparisons of data were per-
`formed: between baseline and while on dexme-
`detomidine infusion; among groups A, B, and
`C; and between neonates and infants. Baseline
`vital signs were compared with the lowest val-
`ues and baseline ABGs with the lowest pH,
`base excess, PO2, and the highest CO2 levels
`while receiving dexmedetomidine. Categorical
`
`Table 2. Baseline patient characteristics
`
`variables were reported as frequencies and
`percentages. Continuous variables were re-
`ported as mean ⫾ SD if the distribution was
`normal and as median with range if the dis-
`tribution was not normal. Paired Student’s t
`test or the Wilcoxon test, depending on the
`distribution of the data being analyzed, was
`used to compare continuous variables before
`and during dexmedetomidine infusion.
`Groups A, B, and C, were analyzed by the
`analysis of variance test or Kruskal-Wallis test,
`depending on the distribution, followed by the
`Bonferroni equation or the Dwass-Steel-
`Chritchlow-Fligner test, respectively, for be-
`tween-group differences. Pain and sedation
`scores were analyzed by analysis of variance
`based on Kruskal-Wallis. Data from neonates
`and infants were either analyzed by an unpaired
`Student’s t test or the Mann-Whitney test. Pear-
`son’s correlation coefficient (r) was used to iden-
`tify any correlation between dexmedetomidine
`dose and vital signs. All reported p values were
`two-tailed, and values of ⱕ.05 were considered
`statistically significant.
`
`RESULTS
`
`A total of 80 patients (39 males and 41
`females) were identified (Tables 1 and 2)
`and divided into three groups: A, n ⫽ 20;
`B, n ⫽ 38; C, and n ⫽ 22. Four of these
`patients were included in our previous
`report (2). Overall there were 1999 indi-
`vidual hourly points of data collected.
`Average age and weight were 4.1 ⫾ 3.1
`months and 5.5 ⫾ 2 kg, respectively.
`Group B had a higher weight compared
`with group C (p ⫽ .01). Fourteen patients
`(17.5%) were neonates with a mean age
`and weight of 15 ⫾ 9 days and 3.2 ⫾ 0.9
`kg, respectively. Seventy-eight surgical
`procedures were adjusted for severity ac-
`cording to RACHS-1 classification and
`the median severity score was 2 (range,
`
`Age (mo)
`Weight (kg)
`Gender (M, F)
`Mechanically ventilated,
`n (%)
`On admission
`At 48 hrs
`Vital signs
`HR (bpm)
`SBP (mm Hg)
`RR (breaths/min)
`CICU length of stay (d)
`
`Group A
`(n ⫽ 20)
`
`3.9 ⫾ 3
`5.5 ⫾ 2
`9, 11
`
`8 (40)
`3 (15)
`
`149 ⫾ 19
`85 ⫾ 12
`32 ⫾ 13
`4 (1–126)
`
`Group B
`(n ⫽ 38)
`
`4.8 ⫾ 3
`6.1 ⫾ 1.9
`18, 20
`
`11 (29)
`3 (8)
`
`146 ⫾ 23
`88 ⫾ 15
`32 ⫾ 12
`3 (1–87)
`
`Group C
`(n ⫽ 22)
`
`3.2 ⫾ 3
`4.4 ⫾ 2
`15, 7
`
`18 (82)
`7 (32)
`
`152 ⫾ 24
`82 ⫾ 18
`29 ⫾ 12
`8 (2–74)
`
`p
`
`.12
`.01a
`
`.52
`.03b
`.7
`.005c
`
`CICU, cardiac intensive care unit; HR, heart rate; RR, respiratory rate; SBP, systolic blood pressure.
`ap ⫽ .002 group B vs. C; bp ⫽ .046 group A vs. B and p ⫽ .017 group B vs. C; cp ⫽ .002 group B vs. C.
`
`655
`
`Petition for Inter Partes Review of US 8,338,470
`Amneal Pharmaceuticals LLC – Exhibit 1045 – Page 655
`
`

`
`1– 6). The total CICU length of stay was a
`median of 4 days (1–126). Group B had a
`shorter CICU stay when compared with
`group C (p ⫽ .002) (Table 2).
`Dexmedetomidine Dosing and Dura-
`tion. Dexmedetomidine was started at a
`mean of 4 ⫾ 6 hours after arrival from
`the operating room. A total of 24 patients
`(30%) received a 0.5–1 ␮g/kg loading
`dose at a rate of 0.1 ␮g䡠kg⫺1䡠min⫺1. The
`mean dexmedetomidine starting dose was
`0.47 ⫾ 0.29 ␮g䡠kg⫺1䡠hr⫺1 followed by a
`maintenance infusion of 0.66 ⫾ 0.26
`␮g䡠kg⫺1䡠hr⫺1. The duration of infusion
`was 25 ⫾ 13 hrs.
`Dexmedetomidine dose requirement be-
`tween mechanically ventilated and nonme-
`chanically ventilated patients did not differ,
`0.62 ⫾ 0.26 ␮g䡠kg⫺1䡠hr⫺1 vs. 0.68 ⫾ 0.26
`␮g䡠kg⫺1䡠hr⫺1 (p ⫽ .3). Similarly, there was
`no difference in dexmedetomidine dose be-
`tween patients who received fentanyl infu-
`sion (group C) and those who did not
`(groups A and B): 0.6 ⫾ 0.21 ␮g䡠kg⫺1䡠hr⫺1
`vs. 0.68 ⫾ 0.28 ␮g䡠kg⫺1䡠hr⫺1 (p ⫽ .2) (Table
`3). Dexmedetomidine duration, however, was
`longer in group C compared with A (p ⫽ .03)
`and compared with B (p ⫽ .002).
`To assess if cumulative experience
`with the use of dexmedetomidine could
`have resulted in higher doses, we com-
`pared the dexmedetomidine dosage be-
`tween the first and the last 40 patients.
`Both starting and maintenance doses
`were significantly higher in the last 40
`patients. Starting dose was 0.34 ⫾ 0.18
`vs. 0.6 ⫾ 0.32 ␮g䡠kg⫺1䡠hr⫺1 (p ⬍ .001)
`and maintenance dose was 0.54 ⫾ 0.2
`␮g䡠kg⫺1䡠hr⫺1 vs. 0.78 ⫾ 0.26 ␮g䡠kg⫺1䡠hr⫺1
`(p ⬍ .001). Furthermore, we analyzed the
`amount of times dexmedetomidine was ad-
`ministered above the recommended adult
`maximum dose of 0.7 ␮g䡠kg⫺1䡠hr⫺1. There
`were a total of 789 recorded doses (39% of
`dexmedetomidine infusion time) above the
`maximum, the majority of which (⬎75%)
`were in the last 40 patients, 169 vs. 620.
`Rescue Sedative/Analgesic Agents.
`Groups A and B, which comprised 73% of
`patients, received dexmedetomidine
`without any other continuous analgesic
`or sedative infusion. Group C had an av-
`erage fentanyl
`infusion dose of 1.7
`␮g䡠kg⫺1䡠hr⫺1 and it continued for 18 ⫾
`15 hrs. Table 3 shows in detail the re-
`quirements of these three groups. There
`was no significant difference in rescue
`sedation/analgesia between patients who
`received fentanyl infusion and patients
`who did not, 2.8 vs. 2.6 doses per 24
`hours of dexmedetomidine infusion, re-
`spectively. Overall, fentanyl (40%), mor-
`
`656
`
`Table 3. Descriptive data on sedatives, analgesics, and hemodynamic infusions among groups A, B, and
`C during dexmedetomidine infusion
`
`Dexmedetomidine
`Dose (␮g/kg/hr)
`Duration/patient (hrs)
`Duration (total patient hrs)
`Fentanyl (␮g/kg/hr)
`Rescue dosesb (doses/24 DEX
`infusion hrs)
`Analgesics (n)
`Sedatives (n)
`Milrinone, n (%)
`Dose (␮g/kg/min)
`Duration (hrs)
`Dopamine, n (%)
`Dose (␮g/kg/min)
`Duration (hrs)
`Epinephrine, n (%)
`Dose (␮g/kg/min)
`Duration (hrs)
`Nitroprusside, n (%)
`Dose (␮g/kg/min)
`Duration (hrs)
`Esmolol, n (%)
`Dose (␮g/kg/min)
`Duration (hrs)
`
`Group A
`(n ⫽ 20)
`
`Group B
`(n ⫽ 38)
`
`Group C
`(n ⫽ 22)
`
`0.72 ⫾ 0.31
`24 ⫾ 12
`471
`
`16 (80)
`0.91 ⫾ 0.24
`23 ⫾ 11
`2 (10)
`2.9 ⫾ 3.1
`5 ⫾ 6
`0
`
`10 (50)
`2.2 ⫾ 1
`18 ⫾ 10
`0
`
`0.65 ⫾ 0.26
`22 ⫾ 11
`820
`
`143 (2.6)
`
`106
`37
`30 (79)
`0.86 ⫾ 0.24
`24 ⫾ 11
`8 (21)
`3.3 ⫾ 0.8
`13 ⫾ 9
`1 (2)
`0.04
`14
`30 (71)
`2.2 ⫾ 1
`12 ⫾ 7
`3 (7)
`157 ⫾ 42
`14 ⫾ 7
`
`0.6 ⫾ 0.21
`32 ⫾ 15
`708
`1.7 ⫾ 1
`84 (2.8)
`
`46
`38
`20 (91)
`0.84 ⫾ 0.26
`32 ⫾ 15
`6 (27)
`3.7 ⫾ 1.2
`16 ⫾ 8
`2 (9)
`0.03 ⫾ 0.02
`26 ⫾ 20
`7 (32)
`1.6 ⫾ 0.7
`16 ⫾ 7
`0
`
`p
`
`.37
`.009a
`
`.68
`.06
`
`.9
`.27
`
`.37
`.16
`
`DEX, dexmedetomidine.
`ap ⫽ .03 group A vs. C and p ⫽ 0.002 group B vs. C; bgroup A and group B rescue doses are
`presented as one group; Values are presented as mean ⫾ SD.
`
`Table 4. Sedation and pain scores
`
`Group A (n ⫽ 20)
`
`Group B (n ⫽ 38)
`
`Group C (n ⫽ 22)
`
`Sedation score (0–3)
`Pain score (0–10)
`
`1.0 ⫾ 0.4
`1.7 ⫾ 0.9
`
`1.2 ⫾ 0.5
`2.0 ⫾ 1
`
`1.4 ⫾ 0.4
`1.9 ⫾ 0.7
`
`p
`
`.28
`.62
`
`Values are presented as mean ⫾ SD.
`
`phine (20%), and chloral hydrate (15%)
`were the most common rescue drugs
`used. Midazolam,
`lorazepam, and ket-
`amine were used less frequently.
`Sedation/Analgesia. The overall mean
`sedation score was 1.2 ⫾ 0.5. Normal
`sleep, defined as easily arousable patient
`without drowsiness, to moderate sedation
`was documented 94% of the time. In re-
`gards to analgesia, the mean pain score
`was 1.9 ⫾ 0.9 and no pain to mild pain
`was documented 90% of the time. Among
`groups A, B, and C, sedation and pain
`scores were similar (Table 4).
`Cardiovascular Effects. Average SBP
`and HR were statistically lower after
`dexmedetomidine was initiated. Blood
`pressure decreased from 89 ⫾ 15 mm Hg
`at baseline, to an average of 85 ⫾ 11 mm
`Hg (5% decrease, p ⫽ .006). HR de-
`creased from 149 ⫾ 22 bpm to 129 ⫾ 16
`bpm (13% decrease, p ⬍ .001) (Fig. 1).
`Further analysis between baseline and av-
`
`erage lowest values showed a decline in
`the SBP to 69 ⫾ 11 mm Hg (22% de-
`crease) and HR to 113 ⫾ 15 bpm (24%
`decrease). Changes in dexmedetomidine
`dose correlated negatively with HR (r ⫽
`⫺.65, p ⬍ .001), and had no correlation
`with SBP (r ⫽ ⫺.2, p ⫽ .22).
`Because of lack of data and because
`the timing of starting dexmedetomidine
`was not uniform in all patients after sur-
`gery, it was difficult to differentiate if
`hemodynamic changes were secondary to
`a potential presence of low cardiac output
`syndrome (LCOS) or dexmedetomidine.
`Nonetheless, the HR trend and the corre-
`lation with dexmedetomidine dosage did
`not support a worsening LCOS because of
`dexmedetomidine (Fig. 1). As noted
`above, the overall HR was lower after
`initiating dexmedetomidine, in contrast
`to the expected higher HR seen in LCOS
`state (7). Additionally, we noticed that
`during the first 8 hrs, when dexmedeto-
`
`Pediatr Crit Care Med 2009 Vol. 10, No. 6
`
`Petition for Inter Partes Review of US 8,338,470
`Amneal Pharmaceuticals LLC – Exhibit 1045 – Page 656
`
`

`
`failed extubation while receiving dexme-
`detomidine. Three had complex CICU
`courses, with chronic lung disease and
`previous prolonged intubation, and two
`were found to have hemidiaphragm pa-
`ralysis that needed surgical intervention.
`Among the patients who were not in-
`tubated at the initiation of dexmedetomi-
`dine, none required intubation during
`the study period. None of the patients had
`any documented episodes of apnea.
`There was no significant change be-
`tween baseline and overall mean RR, 31 ⫾
`12 breaths/min to 31 ⫾ 9 breaths/min (p ⫽
`.83). However, a comparison between base-
`line and the lowest mean RR, 20 ⫾ 6
`breaths/min [35% drop] showed a signifi-
`cant difference (p ⬍ .001). There were 31
`patients with 108 recorded episodes consis-
`tent with low RR for age (5% incidence).
`When we analyzed dexmedetomidine
`dose in relation to the RR, we did not
`find any significant correlation (r ⫽
`⫺.05, p ⫽ .76).
`ABGs were statistically different when
`baseline values were compared with the
`most abnormal ABGs while on dexme-
`detomidine. These changes included
`lower pH level, 7.40 ⫾ 0.1 to 7.37 ⫾ 0.1
`(p ⫽ .006), mild CO2 retention, 45 ⫾ 8 to
`50 ⫾ 12 mm Hg (p ⫽ .001), and lower
`PO2 levels, 91 ⫾ 67 mm Hg to 66 ⫾ 29
`mm Hg (p ⫽ .005). The timing of these
`most abnormal ABGs ranged from 2 to 25
`hrs after starting dexmedetomidine, with
`an average of 4 ⫾ 7 hrs. These ABG
`changes did not differ among groups A, B,
`and C (Table 6).
`Gastrointestinal Effects. Twenty-one pa-
`tients (26%) were fed during the dexme-
`detomidine infusion. One episode of vom-
`iting was documented in one patient.
`Neonates vs. Infants. A subgroup anal-
`ysis was performed comparing neonates
`(n ⫽ 14) with infants (n ⫽ 66) (Table 7).
`Dexmedetomidine dose was statistically
`lower in neonates (p ⫽ .003). Sedation
`and analgesia scores and rescue sedatives
`were similar in both groups. HR and RR
`were statistically higher and SBP was
`lower in neonates.
`Safety and Adverse Events. Two pa-
`tients had changes in the cardiorespira-
`tory variables that may have been attrib-
`uted to dexmedetomidine and thus it was
`discontinued. The first was a 2-mo-old
`patient with aortic coarctation repair who
`had received dexmedetomidine for 8 hrs
`before discontinuation (0.4 ␮g䡠kg⫺1䡠hr⫺1).
`At the time, this patient was simulta-
`neously treated with nitroprusside (2
`␮g䡠kg⫺1䡠min⫺1) for hypertension when a
`
`657
`
`Figure 1. Average heart rate (HR), systolic blood pressure (SBP), and dexmedetomidine (DEX) dose in
`all patients.
`
`Table 5. Hemodynamic variables in groups A, B, and C during dexmedetomidine infusion
`
`Group A (n ⫽ 20)
`
`Group B (n ⫽ 38)
`
`Group C (n ⫽ 22)
`
`SBP (mm Hg)
`HR (bmp)
`RR (br.pm)
`
`85 ⫾ 9
`128 ⫾ 15
`32 ⫾ 8
`
`88 ⫾ 11
`126 ⫾ 13
`30 ⫾ 8
`
`82 ⫾ 11
`136 ⫾ 17
`34 ⫾ 11
`
`p
`
`.19
`.03a
`.34
`
`SBP, systolic blood pressure; HR, heart rate; RR, respiratory rate.
`ap ⫽ 0.01 group B vs. C. Values are presented as mean ⫾ SD.
`
`midine dose increased steadily, there was
`a large negative correlation with HR (r ⫽
`⫺.9, p ⬍ .001). During the 9- to 17-hour
`period, there was no correlation (r ⫽ .04,
`p ⫽ .9); and during the 18- to 28-hour
`period, when there was a steady decline
`in the dexmedetomidine dose, there was
`also a significant negative correlation
`(r ⫽ ⫺.7, p ⫽ .07). These indirect find-
`ings do not support worsening LCOS.
`Figure 1 shows graphically the HR and
`SBP trend in relation to dexmedetomi-
`dine dose and duration. Because vital
`signs were documented only every hour,
`we were not able to detect further
`changes that may have occurred during
`the loading dose or with increased
`dexmedetomidine infusion rate.
`Twenty-seven patients (34%) had at
`least one episode of hypotension and 10
`patients (12.5%) at least one episode of
`bradycardia. Overall, from the 1999 hourly
`recordings, there were 58 hypotensive
`events (3%) and 44 bradycardic events
`(3%). Crystalloid or colloid fluids boluses
`were administered in 23 patients (29%);
`however, we could not determine whether
`these were a part of the postoperative car-
`diac care or if they were associated with
`dexmedetomidine administration.
`
`Pediatr Crit Care Med 2009 Vol. 10, No. 6
`
`Sixty-six patients were on some ino-
`tropic support before dexmedetomidine
`was started. Table 3 shows the inotropic
`agent requirements among groups A, B,
`and C, and Table 5 shows the hemody-
`namic variables. Overall there was no
`substantial difference in the hemody-
`namic infusions, SBP or RR. HR was sta-
`tistically higher in group C compared
`with group B (p ⫽ .01). A total of 50
`patients had significant postoperative sys-
`temic hypertension requiring sodium ni-
`troprusside (47 patients) and esmolol
`(three patients). Thirty-three of these be-
`longed to group B.
`Nine patients had an arrhythmia be-
`fore starting dexmedetomidine. The types
`of arrhythmias included five patients with
`junctional ectopic tachycardia, two with
`first and one with third-degree atrioven-
`tricular block, and one patient with junc-
`tional accelerated rhythm. All arrhyth-
`mias recovered to normal sinus rhythm
`before discharge from the CICU.
`Respiratory Effects. Thirty-seven pa-
`tients (46%) were already intubated at
`the initiation of the dexmedetomidine in-
`fusion. Twenty-four of these were weaned
`off and extubated while receiving dexme-
`detomidine. Five patients subsequently
`
`Petition for Inter Partes Review of US 8,338,470
`Amneal Pharmaceuticals LLC – Exhibit 1045 – Page 657
`
`

`
`Table 6. Comparison of arterial blood gas changes in groups A, B, and C
`
`Group A (n ⫽ 20)
`
`Group B (n ⫽ 38)
`
`Group C (n ⫽ 22)
`
`pH
`Baseline
`Lowest
`CO2, mm Hg
`Baseline
`Highest
`PO2, mm Hg
`Baseline
`Lowest
`Base excess, mmol/L
`Baseline
`Lowest
`
`7.4 ⫾ 0.06
`7.37 ⫾ 0.05
`
`45 ⫾ 6
`54 ⫾ 19
`
`86 ⫾ 54
`73 ⫾ 36
`
`2 ⫾ 4
`1 ⫾ 4
`
`Values are presented as mean ⫾ SD.
`
`Table 7. Neonates vs. infants
`
`7.41 ⫾ 0.06
`7.38 ⫾ 0.05
`
`46 ⫾ 7
`48 ⫾ 8
`
`100 ⫾ 72
`69 ⫾ 26
`
`4 ⫾ 4
`2 ⫾ 4
`
`7.39 ⫾ 0.09
`7.36 ⫾ 0.06
`
`45 ⫾ 11
`50 ⫾ 8
`
`79 ⫾ 71
`55 ⫾ 25
`
`3 ⫾ 6
`1 ⫾ 3
`
`p
`
`.6
`.2
`
`.9
`.4
`
`.1
`.07
`
`.4
`.5
`
`Neonates (n ⫽ 14)
`
`Infants (n ⫽ 66)
`
`p
`
`Group A, B, C (n)
`Dexmedetomidine
`Dose (␮g/kg/hr)a
`Duration (hrs)a
`Rescue sedative/analgesic doses n
`(doses/24 DEX infusion hrs)
`Pain scoreb
`Sedation scoreb
`Vital signs
`HR (bpm)a
`SBP (mm Hg)a
`RR (breaths/min)b
`RACHS–1b
`CICU stay (d)b
`
`3, 7, 4
`
`0.47 ⫾ 21
`27 ⫾ 17
`43 (2.6)
`
`2 (0.8–2.4)
`1.4 (0–2)
`
`140 ⫾ 10
`77 ⫾ 9
`37 (17–55)
`3 (1–4)
`8 (2–30)
`
`17, 15, 34
`
`0.69 ⫾ 25
`25 ⫾ 13
`184 (2.7)
`
`2 (0–4)
`1 (0–2.3)
`
`127 ⫾ 15
`87 ⫾ 10
`28 (18–68)
`2 (1–6)
`4 (1–126)
`
`.003
`.6
`
`.5
`.06
`
`.001
`⬍ .001
`.009
`.1
`.67
`
`DEX, dexmedetomidine; HR, heart rate; SBP, systolic blood pressure; RR, respiratory rate; CICU,
`cardiac intensive care unit; RACHS-1, Risk Adjustment for Congenital Heart Surgery Classification.
`aValues as mean ⫾ SD; bvalues as median with range.
`
`dose of hydralazine was administered. The
`patient’s SBP decreased from a baseline of
`81 to 65 mm Hg, a 19% change, and the
`decision was made to discontinue both the
`nitroprusside and dexmedetomidine infu-
`sions. Blood pressure returned to baseline
`within 15 mins. The second patient was
`also a 2-mo-old with coarctation of the
`aorta repair who had received dexmedeto-
`midine for 11 hrs (0.7 ␮g䡠kg⫺1䡠hr⫺1). The
`patient’s HR had decreased from baseline of
`115 bpm to 89 bpm, a 23% change and
`although normotensive at the time with no
`signs of LCOS, a precautionary decision
`was made by the physician on call to dis-
`continue dexmedetomidine. HR recovered
`to 127 bpm within 1 hr without any further
`interventions.
`
`DISCUSSION
`
`Dexmedetomidine has been increas-
`ingly used in our CICU for sedation, an-
`algesia, and other off-label indications.
`With growing experience, we have ex-
`
`panded its use from adolescents and
`young adults to infants and neonates, and
`to more complex cardiac surgeries. In
`our previous study, although only seven
`patients were younger than 1 yr, we no-
`ticed that this younger population had a
`tendency toward a higher dexmedetomi-
`dine dose requirement (2).
`In this report, which included only
`infants and neonates, the dexmedetomi-
`dine dose was higher compared with our
`previous results in older children, 0.66
`␮g䡠kg⫺1䡠hr⫺1 vs. 0.4 ␮g䡠kg⫺1䡠hr⫺1 (2).
`We also found that with increasing expe-
`rience, the dose had increased to 0.78
`␮g䡠kg⫺1䡠hr⫺1. Although these dose
`ranges are relatively high, they are still
`within the range published in both adult
`and pediatric literature (8 –10). Two pe-
`diatric pharmacokinetic studies by Petroz
`et al (11) and Diaz et al (12), although
`limited by the number of
`infants in-
`cluded, showed that children had similar
`and predictable pharmacokinetics com-
`
`658
`
`pared with adults. In reference to our
`minority neonatal subgroup of patients,
`we found that the dexmedetomidine dose
`was lower compared with the older infant
`population. Given the small number of
`patients, we can only speculate why there
`was such a difference. One possibility is
`that due to the immature neonatal kidney
`function, the renally excreted dexmedeto-
`midine accumulates over time and thus
`the requirement is less. The lower SBP
`and higher RR and HR are likely ex-
`plained by the age difference.
`Dexmedetomidine decreases opioid
`and benzodiazepine requirements, possi-
`bly due to a synergistic or additive effect,
`and in some studies it has been used as a
`single agent for sedation (13, 14). In 25%
`of the patients in this report, dexmedeto-
`midine was administered as the sole sed-
`ative/analgesic agent; approximately half
`of the patients required occasional rescue
`boluses and 27% required an additional
`low-dose fentanyl infusion. A comparison
`between patients who received additional
`fentanyl infusion and those who did not
`showed that there was no difference in
`either sedation or pain score and both
`groups received equal amounts of rescue
`sedative/analgesic boluses. Providing an
`adequate level of analgesia with the least
`amount of side effects is of paramount
`importance, and has been shown to de-
`crease cardiorespiratory morbidity and,
`therefore, hospitalization time. This is
`rather important, taking into consider-
`ation that most CICU patients have sev-
`eral noxious stimuli,
`including chest
`tubes and chest incisions as well as the
`need for mechanical ventilation. Our re-
`sults are in agreement with those of
`Tobias and Berkenbosch (15), who dem-
`onstrated that the use of dexmedetomi-
`dine in pediatric ICU patients was supe-
`rior to midazolam infusion based on
`supplemental rescue dose requirements.
`The study by Tobias and Berkenbosch
`was a small randomized trial, in me-
`chanically ventilated infants and chil-
`dren, and it compared dexmedetomi-
`dine infusion at two different doses, 0.25
`␮g䡠kg⫺1䡠hr⫺1 and 0.5 ␮g䡠kg⫺1䡠hr⫺1 with
`midazolam 0.22 mg䡠kg⫺1䡠hr⫺1. At a dose
`of 0.25 ␮g䡠kg⫺1䡠hr⫺1, dexmedetomidine
`was equivalent to midazolam. At 0.5
`␮g䡠kg⫺1䡠hr⫺1, dexmedetomidine pro-
`vided more effective sedation as demon-
`strated by the need for fewer bolus doses
`of morphine, a decrease in the 24-hr re-
`quirements for supplemental morphine,
`as well as a decrease in the total number
`
`Pediatr Crit Care Med 2009 Vol. 10, No. 6
`
`Petition for Inter Partes Review of US 8,338,470
`Amneal Pharmaceuticals LLC – Exhibit 1045 – Page 658
`
`

`
`of sedation assessment points outside of
`the desired range.
`Dexmedetomidine causes minimal re-
`spiratory depression, making it a poten-
`tially useful agent in nonintubated pa-
`tients. Overall, 95% of the patients in this
`study who were either nonintubated at
`baseline or extubated while receiving
`dexmedetomidine had no clinically sig-
`nificant respiratory compromise. Five pa-
`tients who failed extubation had other
`significant underlying pathologies and
`their failure was not directly attributed to
`dexmedetomidine. Nevertheless, in con-
`trast to our previous study, we did see
`changes in the respiratory variables, and
`although transient, they still warrant at-
`tention. These changes included mild hy-
`percapnia, lower pH, and PO2 levels and
`lower RR. These results are consistent
`with findings from earlier studies by
`other authors, where it was shown that
`both low- and high-dexmedetomidine bo-
`lus, 0.25 ␮g/kg and 2 ␮g/kg, decreased
`resting ventilation and ventilatory re-
`sponse to hypercapnia (16). Other studies
`also demonstrated that dexmedetomidine
`can cause mild decreases in the PO2 levels
`or oxygen saturation, and mild hypercap-
`nia (17). These respiratory changes most
`often are clinically insignificant. How-
`ever, caution is warranted since such al-
`terations can potentially cause significant
`fluctuations in the pulmonary vascular
`resistance and thus change the hemody-
`namic profile in patients with single ven-
`tricle physiology as well as in patients
`with potentially labile pulmonary vascu-
`lar resistance, i.e., after repair of com-
`plete atrioventricular septal defect, trun-
`cus arteriosus, etc.
`The more frequently reported adverse
`events associated with dexmedetomidine
`include a dose-related hypotension and
`bradycardia (13, 17, 18). In this study, the
`prevalence of these side effects was simi-
`lar to that reported in the literature. In
`general, there was only a small drop in
`the SBP and HR, but transiently we saw
`up to 22% and 24% changes, respectively.
`A recent small, prospective study by
`Hammer et al (19) investigated the elec-
`trophysiologic effects of dexmedetomi-
`dine in 12 children who underwent car-
`diac catheterization for possible ablation.
`Although the study was performed in the
`presence of ketamine and propofol, both
`of which may have a negative electro-
`physiologic effect, Hammer et al found
`that dexmedetomidine depressed both si-
`nus and atrioventricular nodal function.
`Dexmedetomidine appears to have a wide
`
`Pediatr Crit Care Med 2009 Vol. 10, No. 6
`
`safety margin; however, its sympatholytic
`properties should be respected and it
`should be used with extreme caution in
`patients at risk for any bradyarrhythmias,
`and in patients who are receiving medi-
`cations that cause vasodilation or have
`negative chronotropic effects (20, 21).
`A less frequent adverse effect de-
`scribed in adults is nausea and

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