Sunteți pe pagina 1din 20

Journal of Intensive

Care Medicine
http://jic.sagepub.com/

Dexmedetomidine and Clonidine: From Second-to-First-Line Sedative Agents in the Critical Care
Setting?
C. Pichot, M. Ghignone and L. Quintin
J Intensive Care Med published online 27 April 2011
DOI: 10.1177/0885066610396815

The online version of this article can be found at:


http://jic.sagepub.com/content/early/2011/04/12/0885066610396815
Published by:
http://www.sagepublications.com

Additional services and information for Journal of Intensive Care Medicine can be found at:
Email Alerts: http://jic.sagepub.com/cgi/alerts
Subscriptions: http://jic.sagepub.com/subscriptions
Reprints: http://www.sagepub.com/journalsReprints.nav
Permissions: http://www.sagepub.com/journalsPermissions.nav

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

Dexmedetomidine and Clonidine: From


Second- to First-Line Sedative Agents
in the Critical Care Setting?

Journal of Intensive Care Medicine


000(00) 1-19
The Author(s) 2011
Reprints and permission:
sagepub.com/journalsPermissions.nav
DOI: 10.1177/0885066610396815
http://jicm.sagepub.com

C. Pichot, MD1, M. Ghignone, MD, FRCPC2, and L. Quintin, MD, PhD3

Abstract
In the critical care setting, a-2 agonists present a multifaceted profile: sedation combined with arousability, suppression of delirium,
preservation of respiratory drive, reduced O2 consumption, preserved renal function, and reduced protein metabolism. In
addition, this review details the reduced arterial impedance, improved left ventricular performance, preserved vascular reactivity
to exogenous amines, preserved cardiac baroreflex reactivity, preserved vasomotor baroreflex activity combined with a lowered
pressure set point: these features may explain the good tolerance observed when a-2 agonists are used as continuous infusion
without any loading dose. Reviewing the literature allows one to suggest that a new management appears possible with arousable
sedation. However, it remains to be demonstrated whether this arousable sedation can be combined with the preservation of
spontaneous ventilation, in the setting of severe respiratory distress, as opposed to conventional controlled mechanical ventilation
combined with conventional sedation. Should such a speculative view be confirmed, then a-2 agonists will move from second-line sedative agents to first-line sedative agents. However, key studies are lacking to demonstrate the effect of a-2 agonists on physiological
endpoints and outcome. Presently, the existing body of data suggests a niche for the use of a-2 agonists in the critical care setting.
Keywords
clonidine, dexmedetomidine, critical care medicine, intensive care medicine, sedation, respiratory drive, weaning, O2
consumption, systolic function, vascular reactivity

Introduction
Continuous sedation in the critical care unit (CCU) presents several drawbacks.1 Thus, daily interruption of sedation and new
ventilation techniques reduce the duration of mechanical ventilation and CCU stay.2 Currently, the goals of sedation include
patient comfort and the ability to tolerate the tracheal tube and
cooperate with the staff. Alpha-2 agonists come in at a juncture,
where the new objectives of CCU care include synchronization
of the ventilator to the patient, maintenance of the functioning of
the neuromuscular/ventilatory system early upon ventilatory
support, and minimizing circulatory side effects while balancing
the patients comfort and cognitive abilities.
This review focuses on the following: (a) the recent evolution toward the use of a-2 agonists as first-line sedative agents,
and (b) their unique ventilatory, circulatory, and metabolic profiles, in addition to their sedative properties. This review is
based on PubMed searches with the following key words: clonidine, dexmedetomidine, sedation, critical care medicine,
intensive care medicine, ventilation, circulation, hemodynamics, and head injury. The articles were considered for their
double-blind, randomized prospective design. Unfortunately,
such articles are few: this literature reflects a succession of
serendipitous findings and the absence of any thoroughly

thought-out pharmacological development. Thus, this patchy literature makes the use of case reports and open studies a necessity
when indications, routes, and dosages are considered. Given the
absence of key physiological and epidemiological studies, this
review is not a comprehensive review but is instead an overview
of the existing data, and inferences are strictly separated from the
data. Some data pertaining to the basic science or the anesthesia
settings are included where relevant. Our institutional protocol for
the use of a-2 agonists is included as a supplement.

Effects on the Central Nervous System and


CCU Applications
The pharmacodynamic profiles of clonidine and dexmedetomidine are almost identical. Sedation, analgesia, and hypotension
1

Critical Care Unit, Memorial Hospital, St Lo, France


Critical Care Unit, Columbia Hospital, WPalm Beach, FL, USA
3
Physiology (CNRS UMR 5123), University of Lyon, Lyon, France
2

Corresponding Author:
L. Quintin, Physiology (CNRS UMR 5123), 8 Rue R Dubois, 69 622 LyonVilleurbanne cx, France
Email: quintin@univ-lyon1.fr

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

Journal of Intensive Care Medicine 000(00)

are mediated via a-2A receptors.3 Dexmedetomidine is more


selective for a-2A receptors (clonidine: 200:1; dexmedetomidine, 1600:1). This selectivity is relative: both drugs evoke
hypertension upon bolus injections, presumably via postsynaptic a-1 receptors (see below) or postsynaptic a-2 receptors.4
Dexmedetomidine produces a larger cardiac parasympathetic
activation (ie, bradycardia), which is of little concern in the
CCU as long as no loading dose is used. Given a nonloading
dose regimen,5-7 a head-to-head comparison is needed to assess
whether a short-acting drug (dexmedetomidine) will minimize
untoward side effects. Conversely, a longer-acting drug (clonidine) may be appropriate in the setting of a CCU stay lasting
days or weeks.

locus coeruleus
Sedation
nucleus ambiguus

rostral ventrolateral medulla


IML
Active bradycardia

PNS heart

SNS

Passive vasodilatation
Passive bradycardia

Sedation
Mechanism. In animals, a-2 agonists are believed to act
through a-2 adrenoceptors, which are located on noradrenergic
(NA) locus coeruleus (LC) cell bodies or dendrites, to inhibit
the release of NA through a negative feedback mechanism
(presynaptic receptors also called autoreceptors)8-10 (Figures 1 and 2). However, functional inactivation of NA terminals
by a neurotoxin (DSP4) or depletion of catecholamine stores
(a-methylparatyrosine, reserpine) do not modify the reduction
in the minimum alveolar concentration (MAC) of halothane that
is evoked by dexmedetomidine.17 Thus, the sedative effect of a-2
agonists may be independent of its action on NA cortical postsynaptic terminals or on NA pontine presynaptic cells bodies.
The arousable sedation observed in the CCU may be accounted
for a low-baseline (tonic) activity and intact (phasic) reactivity of LC neurons, following administration of clonidine15
(Figure 3). In humans, circulatory effects of a-2 agonists are independent of any sedative effect: when a benzodiazepine, nitrazepam, induces more sedation than clonidine, no circulatory
effect is observed.18
Clonidine was synthesized in 1962 as a nasal vasoconstrictor
and the discovery of its sedative and hypotensive effects was serendipitous.19 During the 70s, cardiologists used clonidine as an
antihypertensive, most often at bedtime because of its sedative
effects. This prescription schema blunts the morning increase
in blood pressure.20 Doses ranged from 150 to 1200 mg per oral
(po) in combination with other drugs or up to 3600 mg po when
used as a single antihypertensive agent.21 The drowsiness
recedes in 70% of the patients after 3 or 4 weeks of administration21 with no major evidence of sleep-wake cycle disruption.
Similarly, chronic high doses of clonidine (5400 and 6000 mg/
day po) were not associated with sedation.22 In volunteers,
low-dose clonidine (25 mg) increased rapid-eye-movement
(REM) sleep and decreased non-REM sleep. In contrast,
medium-dose clonidine (150 mg) decreased REM sleep and
increased non-REM sleep.23 Dexmedetomidine induced sedation that closely resembled non-REM sleep in humans.24
In the CCU, clonidine was introduced as an alternative to
chlormethiazol25,26 in the treatment of delirium tremens
(0.45-3 mg/day) or as a supplement (1.4 mg/day) to a
midazolam-fentanyl regimen27 (Table 1).

Figure 1. Impact of a-2 agonists on the noradrenergic dorsal bundle,


catecholaminergic ventral bundle, and cardiac vagal motoneurons:
(a) The a-2 agonists are believed to act through the noradrenergic cell
bodies of the locus coeruleus (LC), a tightly packed nucleus located in
the medial dorsal pons. The sedative effects of a-2 agonists are believed
to be generated through the locus coeruleus (see text). (b) The a-2 agonists inhibit adrenergic presympathetic neurons located in the rostral
ventrolateral medulla (vasomotor center). They do not affect glutamatergic presympathetic neurons: this is in line with the preserved reactivity of the sympathetic system (SNS) following a-2 agonists, despite a
lower set-point.11,12 Adrenergic and glutamatergic presympathetic neurons project onto the intermediolateral cell column (IML), within the
spinal cord, where preganglionic cell bodies are located. Thus, a-2 agonists exert inhibitory effect on the cardiac (slow passive bradycardia)
and vasomotor (slow passive vasodilatation) sympathetic nervous system. (c) Alpha-2 agonists stimulate cardiac vagal motoneurons located
in the nucleus ambiguus external formation (dorsal and medial to the
rostral ventrolateral medulla). They lead to increased cardiac parasympathetic activity (PNS) and increased slope of the cardiac baroreflex,13,14
for example upon brisk increase in pressure (fast active bradycardia).
The combination of increased parasympathetic cardiac baroreflex activity and of inhibited cardiac and vascular sympathetic activity explains the
circulatory properties of a-2 agonists.

Critical care unit delirium. Clonidine suppressed opiate32 and


alcohol25,27 withdrawal. Therefore, its use in the CCU
appears logical; clonidine alone (up to 2.5 mg/day) abated
withdrawal from sedative agents upon weaning from mechanical ventilation.33 Upon tetanus, clonidine (1.2 mg/day then
0.5 mg/day24; or 0.9 mg/day35) supplemented sedation and
magnesium. German intensivists use clonidine liberally as
follows: (a) 35%, 48%, and 56% of the German CCUs use
clonidine for sedation lasting <24, 48, and 72 hours, respectively; (b) clonidine is used during weaning from ventilation
in 62% of German CCUs36; and (c) clonidine is administered
in up to 90% of patients.37 In the late 90s, dexmedetomidine
was introduced as a sedative that was restricted to the
24-hour postoperative period. Following repair of type-A
aortic dissection (in a prospective randomized double-blind
trial, n 15 " 2; clonidine bolus of 0.5 mg/kg followed
by 1-2 mg/kg per hour), the incidence of delirium was similar
between groups but with a lower severity and duration and a

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

Pichot et al

ketamine
NMDA

++

morphine

diazepam

NA
NA

GABA

clonidine

++
1
chlorpromazine, droperidol

Figure 2. Schematic overview of a noradrenergic (NA) synapse:


presynaptic a-2 autoreceptors located on NA cell bodies, terminals
or dendrites, lead to inhibition of NA cell bodies in the locus coeruleus and inhibition of NA release. The schema holds for sympathetic nerve endings. Thus a-1 postsynaptic receptors are not
excited, either within the synapse or at the vascular level. m-Receptors and GABA receptors inhibit NA cell bodies and NA release: a
synergy exists between opiates, benzodiazepines, and a-2 agonists
to inhibit the NA system. This explains why opiates and benzodiazepines requirements are reduced when a-2 agonists are used as
second-line agents. Conversely, when a-2 agonists are used as firstline agents, opiates or benzodiazepines are to be used at reduced
dosage (#50% to #80%; see text). Glutamatergic projections from the
prepositus hypoglossi (nucleus tractus solitarius) and rostralventrolateral medulla (vasomotor center) activate the locus coeruleus through
NMDA receptors15,16: ketamine acts as an inhibitor on these excitatory
projections. By acting on postsynaptic a-1 receptors, neuroleptics act
synergistically with a-2 agonists. In the CCU, such interaction may be
useful upon delirium tremens or drug withdrawal, when high-dose a2 agonists are used as first-line agents and supplemented with
neuroleptics.

shortened CCU stay in the clonidine group.38 Accordingly,


CCU delirium is reduced by approximately 30% (n 103,
2 centers; lorazepam group: 92% vs dexmedetomidine:
63%; P < .01).7 Agitation was less in the dexmedetomidine
group, in which patients were closer to their Richmond Agitation Sedation Scale target score (P < .04).7 These findings
were replicated in a multicenter trial (n 297).6 Such benefits are also observed when dexmedetomidine is compared
with propofol or midazolam in the postcardiac surgery setting (open randomized study, n 30"3 patients, dexmedetomidine: incidence of delirium: 3%; propofol: 50%;
midazolam: 50%).39

Figure 3. Alpha-2 agonists lead to arousable sedation in the critical


care setting. Middle panel: Upon emergence from halothane anesthesia in paralyzed rats, high phasic activity is observed in the LC. (Contrast a nearly orderly pattern of discharge under anesthesia
comprising primarily tonic activity with little phasic activity [A] and a
pattern under emergence comprising only phasic activity [B]): When
clonidine is administered, the tonic activity is entirely suppressed, but
the phasic activity appears untouched [B] (modified from15). [B])
Lower panel: A glutamergic antagonist, kynurenic acid (Kyn) microinjected in the LC suppresses the phasic reactivity preserved after clonidine administration (modified from15). The prepositus hypoglossi
and paragigantocellularis nucleus project onto the locus coeruleus and
provides phasic, glutamatergic, input to the LC.16Could this histochemical organization16 and pharmacological observations15 observed
in the rat be translated to the sedation observed in the critical care?
under a-2 agonist, when the patient is undisturbed, the patient rests
calmly (no tonic LC activity). By contrast upon stimulus, arousability
is preserved (intact high phasic LC activity).

Reduction in opiates and sedative requirements. Adding a-2


agonists to an existing sedative regimen leads to a large reduction in opiates (50%40 and 75%41) and benzodiazepine requirements (80%40). A rigorous design (60 < Bispectral index
[BIS]<70 upon ventilation; 65 < BIS < 95 upon weaning;
85 < BIS < 95 after extubation) led to a 43% and 73% reduction
of propofol supplementation during ventilation and weaning,
respectively. There was a reduction of 58% in morphine

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

flunitrazepamclonidine
(n54) vs. chlormethiazolhaloperidol (n50)
vs. flunitrazepamhaloperidol (n55)
30

randomized; clonidine17 vs.


no-clonidine10

total number of patients20

dexmedetomidine15 vs.
placebo15

clonidine

clonidine

clonidine

clonidine then
1
dexmedetomidine

dexmedetomidine47 vs.
placebo51

clonidine

dexmedetomidine

dexmedetomidine

dexmedetomidine

weaning from
mechanical
ventilation
tetanus

tetanus

tetanus

postoperative
ventilation
and
sedation
postoperative
ventilation
and
sedation
postoperative
ventilation
and
sedation

clonidine

delirium
tremens
delirium
tremens
delirium
tremens

clonidine

clonidine20 vs.
chlormethiazol
haloperidol19
1

clonidine

delirium
tremens

drug

setting

Table 1. Sedative Effects of Alpha-2 Agonists: Overview

thoracic epidural

60-180 mg.h-1*2 days

adjunct : magnesium, morphine,


midazolam up to 70 mg.h-1
adjunct : diazepam 20 mg.h-1; mida- not specified
zolam 30 mg.h-1; morphine 30
mg.h-1; baclofen 75 mg/day,
intermittent vecorunium

clonidine 50 mg.h-1 then dexmedetomidine 0.4


mg.kg-1.h-1

FreshwaterTurner, 2007

Sutton, 1990

Gregorakos,
1997

Liatsi, 2009

Spiess, 1996

Yam,1992

Boher, 1990

Metz, 1980

reference

(continued)

stand-alone vs. placebo; rescue mid- reduction of midazolam (-79%) and Venn, 1999
azolam bolus 0.02 mg.kg-1 then
morphine (-48%); lower respira0.01-0.2 mg.kg-1.h-1 if needed to
tory rate (mean : 14 vs. 21 breaths
Ramsay>2
per min); higher SaO2 (98 vs 96 %)
2.5 mg.kg-1.h-1
0.2-2.5 mg.kg-1.h-1 to Ramsay>2 stand-alone vs propofol 1-3 mg.kg- reduction of alfentanil (-68%);
Venn, 2001
1.h-1, alfentanil 0.25-1.0 mg.kgincreased PaO2/FiO2; increased
1.min-1 to Ramsay>2; no rescue
urine output
medication
6.0 mg.kg-1.h-1*10 min 0.1-0.7 mg.kg-1.h-1 to
stand-alone vs. placebo; rescue pro- reduced propofol (-35%) and
Triltsch, 2002
60<BIS<70 during mechanical
pofol 0.2 mg.kg-1 then 0.5-4
morphine (-63% before extubation;
ventilation, 65<BIS<95 during
mg.kg-1.h-1 to BIS
-100% after extubation) requireweaning, 85<BIS<95 after
ments; difference in norepinephrextubation
ine/dopamine requirements: ns

non-clonidine group : 5 out 10


patients died; clonidine group : 2
out of 17 patients died (p0.04).
recovery

900 mg/day

6 mg/kg/day

stand-alone following previous


weaning from remifentanil
prpofol or fentanylmidazolam
adjunct : diazepam 50 mg/day

increased incidence of pneumonia


and increased duration of
mechanical
ventilation in the chlormethiazolhaloperidol group
positive effect in 25 out of
30 patients

recovery

recovery

reduced duration of
delirium and CCU stay

effect

1.8-2.5 mg/day

up to 1.2 mg until sympathetic


hyperactivity was blocked

associated with fentanyl-midazolam

stand-alone vs. chlormethiazol/


haloperidol

adjunct vs. stand-alone

1.44 mg/70 kg/day

0.45-3.16 mg (mean : 1.11)

maintenance dose

1 mg.kg-1 over 10 min 0.2-0.7 mg.kg-1.h-1 up to


Ramsay>2 whilst intubated

0.9 mg/5 min

0.15-1.2 mg

loading dose

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

dexmedetomidine52 vs.
lorazepam51

dexmedetomidine244 vs.
midazolam122

dexmedetomidine

dexmedetomidine

long-term
ventilation
(up to 120
h)
long-term
ventilation

n20

dexmedetomidine

dexmedetomidine38 vs.
standard of care41

12

dexmedetomidine

dexmedetomidine

38; spontaneous
ventilation33 vs.
mechanical ventilation5

dexmedetomidine

long-term
ventilation

dexmedetomidine190 vs.
saline191

dexmedetomidine

postoperative
ventilation
and
sedation
postoperative
ventilation
and sedation in
children
long-term
ventilation
in medical
CCU
long-term
ventilation

drug

setting

Table 1 (continued)
maintenance dose

adjunct vs. stand-alone

up to 1 mg.kg-1 to 8% 0.8 mg.kg-1.h-1 to Richmond


of the
scale -2 to 1
patients

0.15-1.5 mg.kg-1.h-1

0.25-1.4 mg.kg-1.h-1

0.8 mg.kg-1.h-1*1h

no loading dose

0.2-0.7 mg.kg-1.h-1 to
2<Ramsay<4

no loading dose

reference

reduction in propofol (-87 %)


Martin, 2003
requirements; increased calmness,
tolerance to endotracheal tube,
ease to communicate
absence of respiratory depression
Chrysostomou,
2006

effect

ceiling effect for sedation>1.5 mg.kg- Venn, 2003


1.h-1; reduced adrenaline
requirements; reduced creatinine
(p0.03)
stand-alone; rescue midazolam 1 mg median duration of artificial airway: Shehabi, 2004
or morphine 2 mg/fentanyl 20 mg
7.7 days; 4 out 5 patients present a
reduction in noradrenaline
requirements
dexmedetomidinepropofol vs.
duration of mechanical ventilation
Ruokonen, 2009
placeboplacebo; rescue
shorter (p0.025); dexmedetopropofol 2.4 mg.kg-1.h-1*1h then
midine patients more arousable,
0.8-4.0 mg.kg-1.h-1; rescue
cooperative and better able to
midazolam 1-2 mg up to 3/h then
communicate
0.04-0.20 mg.kg-1.h-1; objective
Richmond scale -3 to -4
stand alone vs. lorazepam
more days without delirium or coma Pandharipande,
1-10 mg.h-1 to Richmond scale -3;
in dexmedetomidine patients (7 vs
2007
rescue fentanyl or propofol
3 days;p0.01); improved shortterm and long-term survival (ns)
stand-alone vs. midazolam 0.06
lower incidence of delirium; shorter Riker, 2009
mg.kg-1.h-1; rescue midazolam
duration of endotracheal intubaand fentanyl
tion (3.7 vs 5.6 days, p0.1);
lower incidence of hospitalacquired infections (overall :
p0.02; urinary : p0.02;
pulmonary: p0.07)

0.2-2.5 mg.kg-1.h-1 up to 7 days rescue propofol 0.2 mg.kg-1 then


for Ramsay>2
0.5-4 mg.kg-1.h-1

1 mg.kg-1*10 min

rescue agents : midazolam, lorazepam, fentanyl, etc . . . .

0.1-0.75 mg.kg-1.h-1

no loading dose

1 mg.kg-1 over 10 min 0.2-0.7mg.kg-1.h-1 to Ramsay>3 stand-alone vs. saline; rescue


before extubation, Ramsay>2
propofol 0.2 mg.kg-1 then
after extubation
0.5-4.0 mg.kg-1.h-1

loading dose

Journal of Intensive Care Medicine 000(00)

requirements,28 which appeared more prominent following


extubation.42 A double-blind, randomized, prospective, multicenter study confirmed these findings in the surgical CCU
(n 401 patients).29 Similar findings were reported in the
pediatric surgical CCU.43
Arousable sedation. With a-2 agonists as stand-alone sedative
agents, the patient lies calmly in bed despite the surrounding
stimuli (noise, tracheal tube, ventilator, etc) but is easily aroused
to full consciousness with the ability to communicate and follow
commands. Following the end of stimulation, the patient returns
to his original indifference to the environment (ataraxia). A large
study (n 79) showed that dexmedetomidine-treated patients
were more arousable, cooperative, and better able to communicate their pain than patients under propofol/midazolam.31
Nurses judged the patients treated with dexmedetomidine as
easier to manage and communicate with.29
Alpha-2 agonists are used either as second-line agents
only31 or as first-line agents.6,7 Two issues are relevant. First,
patients in need of moderate sedation may benefit from a-2
agonists as stand-alone sedative agents. This fits with a daily
assessment of the patient and daily trial of spontaneous breathing. In contrast, patients in need of deep sedation close to
general anesthesia (eg, ARDS-affected patients in the prone
position) may need conventional agents. In this respect, a-2
agonists may also be judiciously supplemented with drugs on
a case-by-case basis to achieve deep sedation and spontaneous
breathing (eg, +hydroxyzine + loxapine + ketamine . . . vs
+ propofol + midazolam + opioid analgesics . . . ). Depending on the dose of clonidine (0.2-2 mg/kg per hour,44 ie, 400 mg3.5 mg/70 kg per day), sedation may be deep (eg, upon
mechanical ventilation and hemodialysis, 3 % Ramsay % 4 or
deeper) or light (eg, upon weaning, after extubation or upon
noninvasive ventilation, 2 % Ramsay % 3). Dexmedetomidine
produced sedation with minimal midazolam in 80% of the
patients5 and, when used alone (up to 2.5 mg/kg per hour, ie,
4.2 mg/70 kg per day), allowed adequate sedation in most
patients in the medical CCU setting. A ceiling effect was
observed above 1.5 mg/kg per hour.45 Second, (a) the drowsiness evoked by clonidine recedes in 70% of the patients after
3 weeks of antihypertensive therapy in the cardiology setting.21
Accordingly, an escape phenomenon occurs after days or
weeks of clonidine sedation (Pichot-Quintin, unpublished
data). (b) Agitation and self-extubation are possible when transitioning from existing sedative regimen to dexmedetomidine.46 However, it should be noted that the patient may
demonstrate spontaneous movements or willingness to communicate under a-2 agonists that are used as first-line
agents.
Amnesia. Clonidine can be used in attention deficit/hyperactivity disorder,47 Tourettes syndrome,48 schizophrenia,49 and
Korsakoff syndrome.50 In keeping with these observations with
clonidine, cognitive function appeared better preserved in
patients
treated
with
dexmedetomidine
(ns).7
Dexmedetomidine-treated patients presented with a higher

incidence of amnesia of their CCU stay,29 but a dose-response


curve is lacking. By contrast, patients treated with dexmedetomidine, but not those treated with propofol, recalled the length of
their CCU stay accurately. Discomfort on the ventilator was a
concern, as opposed to the observation performed in propofoltreated patients.41
Inference. Intensivists are transitioning from the use of a-2
agonists as second-line agents (eg, upon weaning36) to their use
as first-line agents6,7 that are selected as soon as the patients need
noninvasive ventilation51 or invasive ventilation. Little is known
about the use of a-2 agonists alone compared with the various
combinations of other sedative agents used in the CCU.39

Sedation in the Setting of Head Injury


Alpha-2 agonists act as sedative agents in the setting of head
injury. In addition, they act also as sympatholytics, upon rapid
increases in intracranial pressure (ICP) and blood pressure. In
patients with head injury, clonidine (2.5 mg/kg intravenous
[IV] over 10 minutes) leads to a stable carotid blood flow and
arteriojugular O2 difference, despite mild hypotension.52
Accordingly, clonidine (150-450 mg/day through nasogastric
tube) was used in patients with head injury without depression
of consciousness level.53 No overall change in ICP was
observed in patients with head injury receiving clonidine (2.5
mg/kg IV over 10 minutes) when aggregated data are considered. However, an increase in ICP and in arteriojugular O2 difference was reported in 3 out of 12 patients.54 By contrast, the
following was observed in patients with head injury or neurosurgical patients: (a) ICP was slightly lowered, but cerebral
perfusion pressure was slightly increased following dexmedetomidine; and (b) hypotension resulted in patients with head
injury following a loading dose of the a-2 agonist.55 Thus, an
IV loading dose should be avoided in head-injury patients, and
cerebral perfusion pressure must be maintained.
In patients with head injury, the Lund concept is applied
to reduce ICP, prevent the breakdown of the blood-brain barrier, and decrease vasogenic edema56 in patients with lownormal cerebral perfusion pressure (60-70 mm Hg). Therapy
consists of low-dose thiopentone, dihydroergotamine, a bblocker (metoprolol), and clonidine (1.5-8 mg/kg per day, ie,
ca 110-560 mg/70 kg per day). In a retrospective study, this
approach improved outcome (deaths: 47% vs 8%; good recovery: 42% vs 72% in the historical [n 38] vs the Lund concept
[n 53] groups, respectively).57 A retrospective study from
another Swedish group58 confirmed the Lund findings. However, no randomized prospective trials in a large sample of
patients have substantiated these findings.59 Thus, the Lund
concept cannot be viewed as a standard therapy.

Analgesia
In animals, clonidine has antinociceptive activity60 and is 5 to
10 times more potent than morphine at identical doses.61
Analgesia appears primarily linked to a supraspinal site of

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

Pichot et al

action.62 In conscious healthy volunteers, although


medetomidine did not change heat, dental, or tourniquetinduced pain, it attenuated the unpleasantness produced by
ischemia. Thus, medetomidine lowered the affectivemotivational component (analgognosia) with little influence on
sensory intensity itself.63 In humans, (a) clonidine reduced the
amount of intraoperative opiates by 45% to 75% with improved
beat-by-beat variability in blood pressure;64-66 (b) combined with
proprofol, epidural clonidine, in a dose-dependent manner,
allowed the performance of intraabdominal surgery.67 Similar
observations were reported with dexmedetomidine at high doses
(5-10 mg/kg per hour IV) without opiates.68 High-dose dexmedetomidine (1.4 mg/kg over 10 minutes, followed by 0.7 mg/kg per
hour69; 10 mg/kg per hour followed by 1 mg/kg per hour with sevoflurane 1%70) induced anesthesia without opiates in morbidly
obese patients. Alpha-2 agonists may not be used as the sole agent
in the CCU when pain is anticipated because medical patients
need a little supplementation (5 mg of morphine/day); by contrast, trauma/surgical or dialyzed patients need larger doses (23
or 63 mg/day, respectively).5
Inference. The core effect of a-2 agonists is to modify the
perception of pain and promote ataraxia. Thus, given an a-2
sedation regimen, 2 alternatives are available if pain is a documented issue: (a) selection of nonopiate analgesics to allow for
spontaneous ventilation (eg, pressure support ventilation)
because of the preserved ventilatory drive (see below) or (b)
initial usage of an approximately 80% reduced opiate dose, followed by titration of the opiates to the desired analgesic effect.

Effect on Ventilation and CCU Applications


Mechanism
In vitro studies on the neonate mouse brain stem revealed that
clonidine increases the respiratory frequency and counteracts
the depression caused by midazolam.71 In halothaneanesthetized rats, (a) clonidine induced a transient 15% to
20% decrease in respiratory rate72 with an increase in tidal volume73 (b) dexmedetomidine did not potentiate the respiratory
depression induced by alfentanil.74 In healthy volunteers, clonidine (300-400 mg po) alone did not depress the slope of the
ventilatory response to CO2 rebreathing or the slope of the
occlusion pressure to CO2. Further, clonidine did not worsen
the respiratory depression induced by morphine (210 mg/kg
intramuscular [IM])75; at a dose of 3 mg/kg IV, it changed neither the minute volume nor the end-tidal CO2 but lowered the
response to CO2 rebreathing,76 mimicking the effect of slowwave sleep. Finally, following clonidine, O2 consumption
(VO2) is reduced in healthy volunteers (#12%) at rest77 and
upon shivering in the postsurgical setting.78
Following upper airway surgery in patients with sleep
apnea, the minimal O2 saturation was higher following clonidine (2 mg/kg po).79 When dexmedetomidine was used as a
total intravenous anesthetic agent, O2 supplementation was
not necessary in some instances.68

Noninvasive Ventilation and Weaning


Clonidine (1-5 mg/kg tid, route unspecified) allowed the adaptation of young children to noninvasive ventilation.80 Clonidine
improved ventilatory parameters (frequency/tidal volume,
PaO2/FIO2, duration of weaning, CCU stay, and severity of delirium) in patients presenting with delirium and recovering from
type-A aortic dissection.38 Following unsuccessful weaning, clonidine (0.9-1.8 mg over ca 20 minutes) reduced VO2 by 45%.33
The respiratory rate was lowered from 22 to 14 breaths/min.33
Accordingly, dexmedetomidine (3 mg/kg per hour over 5 minutes
then 0.2-0.7 mg/kg per hour) administered to patients presenting
with agitation under noninvasive ventilation allowed weaning
of noninvasive ventilation without agitation, reduced respiratory
rate from 29 to 22 breaths/min (n 10), and improved PaO2/
FIO2 and PaCO2.51 In a medical/surgical CCU setting, dexmedetomidine sedated patients for an extended period of time,
following extubation (26 hours postextubation).5
Dexmedetomidine had the following effects in the postsurgical setting: lowered respiratory rates (placebo: 21 breaths/
min vs dexmedetomidine: 14 breaths/min) and increased O2
saturation40 after general surgery with routine ventilation;
reduced morphine requirements (#63%) and improved PaO2/
FIO2 when titrated to Ramsay & 3 before extubation (0.4 mg/
kg per hour) and Ramsay & 2 after extubation (0.17 mg/kg per
hour), as shown by retrospective42 and prospective analysis (n
10 " 2);41 reduced the duration of mechanical ventilation
(dexmedetomidine: 77 hours; propofol/midazolam: 110 hours;
n 79; P .025)31; lowered the incidence of atelectasis (n
401 patients, P .01)29; and increased the PaO2/FIO2 ratio
before and after extubation in the surgical CCU setting.42
Finally, a multicenter trial (n 297) that compared highdose dexmedetomidine (up to 1.4 mg/kg per hour) to midazolam reported a reduced duration of intubation (#1.9 day),
although CCU stay was not shortened.6 In the pediatric cardiac
surgical setting, dexmedetomidine was used in spontaneously
breathing children in the immediate postoperative period without any ventilatory side effects.30

Bronchodilatation
Outside the critical care setting, inhaled clonidine (75 mg)
allows an improvement in forced expiratory volume (FEV1)
and peak expiratory flow rate in asthmatic patients.81 These
beneficial effects were not observed when clonidine was administered orally.82 In dogs, IV dexmedetomidine caused minor
bronchodilatation and blocked the histamine-induced bronchoconstriction, but inhaled dexmedetomidine did not.83 In a 4year-old agitated child in whom inhalational therapy was
impossible, dexmedetomidine allowed one to deal with status
asthmaticus.84

Inference
Easy weaning may be a consequence of the following: (a)
arousable sedation, which allows frequent assessment of the
patient; (b) the absence of depression of the ventilatory drive

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

Journal of Intensive Care Medicine 000(00)

in vitro71 as well as a large reduction in the requirements for


sedative agents and opiates, which further preserves the
respiratory drive; (c) the reduction in emergence delirium following mechanical ventilation under conventional sedation85;
and (d) the reduction in O2 consumption77,78 upon weaning.33
Thus, a-2 agonists allow sedation to be maintained throughout
mechanical ventilation from intubation6 to weaning and noninvasive ventilation. Sedation under spontaneous ventilation is
suitable to the current requirement for daily spontaneous
breathing trials as the standard of care, supports the trend
toward the synchronization of the ventilator to the patient, and
promotes the maintenance of full functionality of the
respiratory-muscular system from intubation to extubation.
However, the use of a-2 agonists remains speculative if initiated from the start of respiratory assistance (eg, for ARDS),
to reduce shivering or VO2 consumption in the setting of ARDS
or intentional hypothermia (eg, in the neuro-CCU).

Effects on Circulation and Side Effects


Mechanism
In animals, cardiac parasympathetic activation by clonidine is
enhanced after depletion of catecholamines by reserpine-amethyparatyrosine.86 Sympathetic nerve activity is inhibited
by clonidine, even after reserpine-a-methylparatyrosine treatment.87 Thus, a-2 agonists may present circulatory effects
entirely independent of adrenergic mechanisms. Despite these
facts, a-2 agonists are believed to act through a-2 adrenoceptors
located on adrenergic cell bodies (autoreceptors) of presympathetic neurons in the vasomotor center11,88 (Figure 1). In vivo
electrophysiological data89 contradict pharmacological data,
which favor an action through imidazole receptors.90 The reactivity of the sympathetic vasomotor reflex persisted after hypotension.11,12,91 In dogs, following severe coronary stenosis, (a)
clonidine (10 mg/kg IV) suppressed the increased poststenotic
coronary resistance and myocardial lactate consumption92; (b)
dexmedetomidine increased the endocardial/epicardial bloodflow ratio, despite a reduction in myocardial blood flow.93
Alpha-2 agonists are known to blunt tachycardia and hypertension, and to reduce plasma catecholamine concentration.
Thus, the delineation of the clinical applicability of these properties is not needed. By contrast, safety issues are key: inotropism, vascular reactivity, and circulatory side effects.

Inotropism
In in vitro rat studies, an unchanged ejection fraction is
observed, compatible with an absence of intrinsic negative inotropism. The in vivo observations may be the end result of an
afterload reduction combined with a reduced inotropic state;
the reduced inotropism is secondary to the reduced cardiac
sympathetic drive to the heart.94 in vivo studies on the patients
with chronic heart failure (CHF) following clonidine (150 mg
bolus) showed reduced left-filling pressure, bradycardia, and
increased stroke volume and cardiac output.95 These observations were repeated as follows: (a) after clonidine 400 mg

po96 or 300 mg/day for 1 week97 or another a-2 agonist, guanabenz (8-12 mg),98 NYHA III to IV patients showed lowered
pulmonary capillary wedge pressure, bradycardia, increased
stroke index, and constant cardiac output. However, systemic
hypotension (BP < 90 mm Hg) was observed in 3 patients96;
and (b) upon arrival in the CCU, clonidine-treated patients who
were recovering from coronary bypass surgery presented with
higher mixed venous saturation.99 More intriguingly, a greater
improvement in sicker patients was observed.97 These latter
observations were repeated as follows: (a) Despite lower exercise capacity before randomization, improvement in the exercise capacity was larger in the clonidine-treated patients100;
and (b) upon acute myocardial infarction associated with
hypertension and high capillary wedge pressure, clonidine
(5 mg/kg IV over 5 minutes) led to minimal bradycardia,
lowered the wedge and systemic pressure while maintaining
cardiac output. The improved left ventricle (LV) function was
more prominent in the patients with higher wedge pressures.101 In patients with hypertension, clonidine (150 mg
bolus) led to intriguing observations: high cardiac output was
converted into normal cardiac output (7.2-4.9 L/min). By
contrast, low cardiac output was converted to normal cardiac
output (2.6-4.2 L/min). An 11% increase in stroke volume
was observed when the HR was lowered more than the cardiac output.102 In patients with hypertension who presented
with a diastolic pressure of >130 mm Hg, clonidine (5 mg/
kg IV over 20-36 minutes) increased stroke volume and ejection fraction from 59% to 71%.102

Vascular Reactivity
In vitro, clonidine inhibits the activation evoked by isoproterenol. By contrast, clonidine led to a 50% to 60% increase in
isoproterenol-evoked activation. The sensitivity of the b-adrenergic receptor appeared to increase, following clonidine.103 In
healthy volunteers, clonidine (2 mg/kg over 10 minutes) evoked
an increase in the density of b-adrenoceptors on lymphocytes,
with a decrease in affinity. A possible externalization of badrenoceptors following lowered plasma catecholamine concentration was hypothesized. This was not observed in vitro.104
In patients with CHF, clonidine (150 mg IV bolus) lowered the
pressure in the isolated vein segment but increased the pressor
response of the isolated vein segment upon deep breathing.105
In supine hypertensive patients, clonidine (200-400 mg/day)
increased vascular reactivity: the dose of phenylephrine necessary to increase the pressure by 25 mm Hg (PE25) was reduced
from 96 to 39 mg. The chronotropic response to isoproterenol
(increase in HR by 20 beats per minute: CD20) was
increased.106 This upregulation of a-1 and b-receptors agrees
with the increased response to noradrenaline observed after
methyldopa administration in patients with hypertension .107
Upon recovery from major surgery and aortic surgery, respectively, the response to phenylephrine108 and isoproterenol was
increased.109 Following liver transplant, intraoperative noradrenaline and postoperative vasoconstrictor and inotropic
requirements were reduced in the clonidine-treated patients.110

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

Pichot et al

Reduced Vasopressor Requirements


In the pediatric cardiac surgical CCU setting, dexmedetomidine did not evoke any increase in inotropic requirement.30
In the medical CCU setting, adrenaline requirements were
reduced, following introduction of dexmedetomidine.45
Similarly, following cardiac surgery, dexmedetomidine
administration was associated with a lower incidence of
administration of adrenaline (randomized open label, inclusion: n 148 dexmedetomidine-treated patients vs 147
propofol-treated patients; 18 dexmedetomidine-treated
patients vs 32 propofol-treated patients requiring adrenaline; P .03).111 Furthermore, vasopressor requirements
were decreased and the duration of their administration was
shortened in septic patients. The 2 groups of patients were
unequal in their incidence of vasopressors administration
at enrollment.112 In a patient with sepsis, increases in
plasma concentrations of endogenous noradrenaline and
adrenaline were preserved following clonidine.113 In a
patient with septic shock (SBP < 70 mm Hg despite NA
3 mg/kg per min), SBP increased to '100 mm Hg within
3 hours of clonidine administration, and vasopressor
requirements were reduced up to 20% of baseline within
48 hours of clonidines introduction.114

Other Side Effects


The baseline sympathetic activity was lowered, but the reactivity of the vascular and cardiac sympathetic systems was unaffected.12 However, bradycardia and hypotension are to be
expected, especially in hypovolemic patients.
Bradycardia was observed in children who presented with
head injury and were treated with hypothermia combined with
dexmedetomidine-remifentanil.115 In this observation, differentiating the bradycardia linked to hypothermia or remifentanil
or dexmedetomidine appears elusive.
No hypotension was observed in infants (0-24 months)
recovering from cardiovascular surgery, but one brief episode of sinus bradycardia resolved spontaneously (retrospective study; n 50; clonidine: 0.18-3.6 mg/kg per hour
IV).116 A similar absence of bradycardia and hypotension
was observed in the setting of a medical pediatric CCU
(respiratory failure; n 24; clonidine 3 mg/kg 3 times per
day through the nasogastric tube). This English pediatric
CCU has used clonidine as a first-line sedative agent in
combination with morphine since 2000. Approximately,
85% of the patients receive clonidine during their CCU
stay,117 which agrees with the German data in adults.37,29
In contrast, a retrospective study (n 121) using dexmedetomidine (0.15-0.70 mg/kg per hour) in pediatric patients
observed hypotension in 16% of the patients and bradycardia in 12% of the patients.118 A loading dose of dexmedetomidine that is administered slower than the established
infusion regimen led to a lower incidence of hypotension,
but bradycardia still occurred.119 Furthermore, while a loading dose of dexmedetomidine was followed by

hypertension, then hypotension,40,29 a slow infusion without


loading dose suppressed almost all hypotensive effects.5
This was also observed in patients with head injury54,55 and
a pediatric cardiac surgical CCU.30 In the medical CCU
setting, volume loading and a reduction in the loading
doseof dexmedetomidine appeared to be relevant.45 Of
note, in the setting of long-term mechanical ventilation,
little6 or no7 loading doses of dexmedetomidine were used.
Overdose of a-2 agonists appeared to be surprisingly
well-tolerated.120,121
Hypertension may be observed with a-2 agonists in the following scenarios: (a) loading with a clonidine bolus or dexmedetomidine rapid infusion; (b) oral administration of high-dose
clonidine (5000-6000 mg/day ie, ca 3-4 mg/kg per hour; plasma
concentrations: 14-26 ng/mL)22; and (c) high-dose dexmedetomidine (4 mg/kg per hour for several hours) in a child presenting with head injury. Decreasing the infusion rate normalized
the pressure.122

Arrhythmias
The parasympathomimetic effect is demonstrated in animals
where clonidine increases the excitability of cardiac vagal
motoneurons13 and enhances bradycardia upon stimulation of
the Bezold-Jarisch reflex.123 In humans, a very small dose of
clonidine (0.2 mg/kg over 5 minutes, ie, 14 mg IV/70 kg) leads
to sinus slowing and depression of nodal conduction, which is
of lesser importance than those observed following b-blocker
administration.124 Lengthening of the PR interval is more prominent when the baseline PR interval is long.124 Clonidine
(450 mg po per day) administered chronically for hypertension
may be associated with dizziness, syncope, and short sinus
arrest (4.5 seconds).125 Clonidine (150 mg IV bolus may lead
to isorhythmic atrioventricular dissociation.95 A 2-minute
asystole has been reported upon sternal retraction following
dexmedetomidine (1 mg/kg over 10 minutes then 0.2 mg/kg per
hour) in a patient with a history of vigorous exercise who was
treated with a parasympathomimetic drug, pyridostigmine.126

Inference
In the CCU, a-2 agonists appear surprisingly well-tolerated,
provided that hypovolemia is avoided before and throughout
administration of a-2 agonists. A very slow infusion of an a2 agonist (eg, dexmedetomidine: 0.4 mg/kg hour for 1 hour
without loading dose followed by 0.2 to 0.7 mg/kg per hour
infusion to a 2 < Ramsay < 45; clonidine: 0.2 to 2 mg/kg per
hour without any loading dose or rapid infusion44) appears
satisfactory. These regimens may be combined with very low
doses of noradrenaline or dobutamine if hypotension or bradycardia occurs.5 This would maintain or transiently increase
the perfusion pressure, depending on the organ at risk (eg,
patients with head injury, high perfusion pressure to maintain
an adequate diuresis . . . ) or HR, if venous saturation is low.

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

10

Journal of Intensive Care Medicine 000(00)

Effects on the Gastro-Intestinal Tract,


Kidney, and Metabolism
Gastrointestinal Tract
The half-life of clonidine appears to be unchanged in cirrhotic
patients (cirrhotic patients: 25.4 + 3.0 hours; normal individual: 20-25 hours).127,128 Clonidine (150 mg po for 7 days)
reduced hepatic venous pressure and gradient. In cirrhotic
patients with elevated noradrenaline (>300 pg/mL) clonidine
(75 mg twice daily for 3 months; double-blind randomized) led
to the following: increased sodium excretion, loss of body
weight, reduced diuretic requirements, no renal impairment,
reduced number of paracenteses, and delayed readmission for
tense ascites.129 Clonidine 2.5 mg/kg IV reduced cardiac output
but did not affect hepatic flow in cirrhotic patients (mostly with
ascites:30 out of 44 patients). A reduced portal venous pressure
is explained by a reduction in postsinusoidal resistance. Despite
a fall in BP, there was an increase in renal flow, glomerular filtration rate, and urine flow.130
Small bowel transit time is prolonged in volunteers (placebo: 90 + 7 minutes; clonidine 148 + 20 minutes, n
6; randomized single blind)131 and in diabetic patients,132 but
emptying of the gall bladder was accelerated.132 Intestinal
pseudoobstruction has been reported following clonidine
administration in the ambulatory setting133 and in the critical
care setting; this was observed in patients treated for delirium
tremens with high-dose clonidine (up to 3000 mg/day) in addition to midazolam and pethidine.134

Kidney
NYHA III to IV patients treated with clonidine (300 mg/day for
1 week) experienced a halving of their dose of diuretics and a
reduction of edema.97 ASA 1 patients treated with clonidine 5
mg/kg presented a larger intraoperative urine output and sodium
and potassium excretion.135 Patients with hypertension (n
17) who underwent coronary surgery with cardiopulmonary
bypass and were premedicated with clonidine (3 mg/kg po) presented no postoperative changes in serum creatinine, as
opposed to placebo patients.136 A similar preservation was
observed in liver transplant110 and in coronary patients (n
48).137 The incidence of elevated urea (BUN) was lowered
by half in dexmedetomidine-treated patients, following cardiac
surgery.111 In the CCU, dexmedetomidine-treated patients presented with a higher urine output (P .12).41 Lower creatinine
concentrations were observed in the medical CCU setting (nonrandomized design; P .02).45

is achieved after 1 to 3 hours, and half of it is recovered,


unchanged in the urine. However, the half-life of the drug is
increased in renal failure.139 Dexmedetomidines terminal
half-life (t1/2b 3.14 hours) does not appear to be modified
in stable patients presenting to the surgical CCU140 as compared to healthy volunteers. The elimination half-life of dexmedetomidine (0.6 mg/kg IV over 10 minutes) was shorter
(113 + 11 vs 136 + 13 minutes) in volunteers with renal
impairment (creatinine clearance: 21 + 9 mL/min, n 5) compared with healthy volunteers (creatinine clearance: 122 + 31
mL/min, n 5). This supports an hepatic elimination process.
The intensity of the sedation was more prominent in the volunteers with renal impairment.138 No data pertaining to the
reduced requirements of clonidine or dexmedetomidine in
acute renal failure are available. As hemodialysis/filtration is
often required upon acute renal failure, the dosage of a-2 agonist has to be adjusted and titrated to the desired effects on
sedation and circulation.

Metabolism
A nonrandomized retrospective analysis showed that protein
catabolism was almost entirely suppressed in alcohol abusers
who were treated with clonidine, following esophageal cancer
resection; additionally, the length of hospital stay was
reduced.141 Clonidine had the following positive effects in various patient groups: increased gastric intramucosal pH after
cardiopulmonary bypass,142 reduced metabolic acidosis following liver graft declamping,110 and decreased plasma lactate
levels 12 hours after cardiac surgery.143

Sepsis
In septic rats, dexmedetomidine and clonidine improved circulation, reduced mortality (94%-44%), decreased cytokine concentrations,144 and inhibited the aggregation of neutrophils in
the lung.145 This was replicated in a dose-related manner, leading to a reduced acidosis and mortality from 84% to 20% and
preserved blood pressure.146 In humans, following (a) cardiac
surgery performed on clonidine patients (1-3 mg/kg per hour for
6 hours: ca 1.2 mg), early proinflammatory response was attenuated143; (b) major surgery, dexmedetomidine lowered interleukin 6 concentrations but had no effect on cortisol and ACTH
concentrations.147

Inference
Kidney function and peripheral blood flow appear well preserved. Response to sepsis appears improved.

Pharmokinetics
The elimination of clonidine and dexmedetomidine is primarily
through the kidney and liver, respectively, with different elimination half-lives in healthy normotensive participants: 20 to 25
hours for clonidine128 and 136 + 13 minutes for dexmedetomidine.138 Clonidine is absorbed easily after oral administration,
and its bioavailability is close to 100%. Its peak concentration

Miscellaneous
Withdrawal
Withdrawal syndrome after discontinuation of a-2 agonists has
been reported following long-term outpatient treatment in the
cardiology setting but not following acute treatment in the

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

Pichot et al

11

toxicology,32 anesthesia, or CCU setting. No withdrawal syndrome was observed after using dexmedetomidine for 3 to 4
days in a multicenter study (n 244).6 However, tapering high
doses of a-2 agonists over 2 to 4 days is advisable, after days or
weeks of administration.

Route
In the United States, given the absence of IV preparation, (a)
some intensivists have used the epidural preparation of clonidine for i.v administration (b) via the transdermal route, 3 to
4 days are needed to reach steady-state plasma concentrations.139 To circumvent this problem, a combination of nasogastric and transdermal routes may be used and work is
needed to achieve a simple and efficacious administration
protocol for clonidine when IV clonidine is not available.
Finally, there will be regulatory problems. In France,
although IV clonidine is cheap and available, its CCU44 use
is off-label. The likelihood of the licensing of dexmedetomidine in the European market may warrant its use, regardless of the cost. In the United States, present regulations
allow the use of dexmedetomidine for 24 hours only, at a
dose of %0.7 mg/kg per hour.148 The FDA granted permission
to use dexmedetomidine at dose twice that high, for up to 30
days of mechanical ventilation,148 presumably to assess
safety within a research protocol.

Outcome
The CCU stay of eclamptic patients was shorter for patients
treated with dexmedetomidine (mean stay 45 hours) than for
those treated with midazolam (mean stay 83 hours).149
Improved outcome was observed in the aortic surgery setting,
where the combined incidence of cardiac death and myocardial
infarction was lowered following aortic surgery and a-2 agonist administration.150 Clonidine improved in-hospital and
long-term survival following elective noncardiac surgery in
coronary patients.151 a-2 agonists reduced mortality and myocardial infarction following vascular surgery152 and decreased
the risk of cardiac death by 52%.153 Clonidine was associated
with better survival in the CCU in patients with tetanus (prospective design; nonclonidine patients [n 10]: death: 50%;
clonidine-treated patients [n 17]: death: 11%; P .04).154
Trauma-infected patients with alcohol withdrawal showed a
higher incidence of pneumonia (P .04) and longer duration
of ventilation (P .03) when treated with chlormethiazol/
haloperidol (n 50) as compared with treatment with flunitrazepam/clonidine (n 54).26 Dexmedetomidine was associated
with a trend toward better outcome and longer survival (n
103).7 When an analysis was restricted to septic patients, dexmedetomidine was associated with a reduction in death rate by
70% (dexmetedomidine: n 19; lorazepam: n 20; P
.04).112 A large (n 297), multicenter trial observed a reduction in infections in dexmedetomidine-treated patients (P<.02;
urinary tract infections: P<.02; hospital-acquired pneumonia:
P .07).6

Cost
Dexmedetomidine and lorazepam treatments cost US$4675
and $2335, respectively (median-calculated cost for the study
drug; total pharmacy cost: dexmedetomidine [n 45]
$27 460 vs lorazepam: [n 45] $20 660, P .15)7 (US market: one 2-mL vial of dexmedetomidine, 100 mg/mL$50-$80;
French veterinary market: 1 mL 500 mg of dexmedetomidine, 10 mL vial, 135). On the other hand, the ICU costs are
similar in the 2 groups.7 The estimated costs (2008) in France
for the sedation of 1 patient for 24 hours with clonidine (1 mg/
kg per hour, ie, 3.4 mg/70 kg per day) as a first-line sole sedative agent and other conventional sedation regimens are as follows: clonidine, 5.1 (French market: 1 vial of clonidine 150
mg in 1 mL 0.42); midazolam sufentanil, 6.4; and propofol remifentanil, 55. Such comparison is financially relevant but may be biased because no head-to-head drug
comparison has been performed, for example on the French
market. Nevertheless, these drug regimens are usually combined (eg, propofol remifentanil vs midazolam sufentanil
vs clonidine alone).

Inference
The studies cited in this review show a trend toward a possible
reduction of morbidity and mortality with the use of a-2 agonists in critically ill patients. However, this is based on small,
nonrandomized or retrospective studies.112,153,154 Only Phase
III or IV randomized controlled trials will be able to fully prove
a change in outcome. Two hypotheses may account for the
improved outcome: (a) improved cardioventilatory interactions lead to accelerated recovery: the length of mechanical
ventilation and the iatrogenic complications are reduced; (b)
a-2 agonists are sympathetic inhibitors and parasympathetic
activators: parasympathetic activation increases resistance to
inflammation.155
Furthermore,
clonidine
stimulates
macrophages.156

Conclusion
Alpha-2 agonists evoke sympathetic inhibition combined with
parasympathetic activation. These various sites of action
explain the multifaceted profile of a-2 agonists: sedation combined with arousability, reduced incidence of delirium, preservation of the respiratory drive, allowing the use of modern
ventilatory techniques (eg pressure support) with reduced
intrathoracic pressure, weaning from mechanical ventilation,
and initiation of noninvasive ventilation, reduced O2 consumption, reduced arterial impedance and improved LV performance, preserved vascular reactivity to exogenous amines,
preserved cardiac baroreflex reactivity, preserved vasomotor
baroreflex activity combined with a lower set point, preserved
renal function, improved tissue perfusion, and reduced protein
metabolism. Hypotension and bradycardia appear manageable
if volemia is repeatedly optimized before and throughout the a2 agonist treatment and slow IV infusion without bolus administration is considered as the only therapeutic modality. It

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

12

Journal of Intensive Care Medicine 000(00)

appears that a new management strategy is possible148,157


Arousable sedation may be combined with spontaneous ventilation (eg, pressure support ventilation). Accelerated/early
weaning and improved outcome need confirmation
As research was carried out primarily in the cardiology,
anesthesiology, and surgical CCU settings, the literature on
a-2 agonists with respect to physiology and outcome in the
medical CCU setting presents limitations. Double-blind randomized prospective studies are needed to substantiate a niche for
a-2 agonists in the CCU.

Declaration of Conflicting Interests


The author(s) declared a potential conflict of interest as follows: L
Quintin received grants and honoraria from Bohringer-Ingelheim
France, UCB Pharma, and Abbott International (1986-1995).

Funding
The author(s) disclosed receipt of the following financial support for
the research and/or authorship of this article: Block grants from University of Lyon-CNRS to UMR 5123, PICS CNRS and PEPS CNRS to
LQ.

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

Pichot et al

13

Institutional protocol for the use of alpha-2 agonists as sedative agents in our CCU
Indications (for clonidine according to French Society of Critical Care Medicine [44]):
Weaning from mechanical ventilation.
Hyperdynamic circulation (orage neurovegetatif).
Emergence from sedation in head-injured patients.
Indications (for clonidine according to French regulatory authority and French compendium of specialties : dictionnaire
Vidal):
Malignant hypertension, hypertensive encephalopathya
Aortic dissectionb.
Left ventricular insufficiency in the setting of acute pulmonary oedema.
Pre-eclampsia (2d line agent).
Indications (for alpha-2 agonists according to the literature or the authors):
Tachycardia and/or hypertension.
Head injury (with preserved cerebral perfusion pressure).
Tetanos.
Delirium Tremens.
Weaning from heroin, cocaine.
Weaning from sedation.
CCU delirium.
Preservation of sleep-wake cycle in the CCU.
VO2 reduction.
Suppression of shivering.
Adaptation to intentional hypothermia.
Adaptation to non-invasive ventilation.
Weaning from mechanical ventilation.
Absolute contra-indication:
Absolute or relative hypovolaemia : Adequate volemia is mandatory before and throughout administration of a-2 agonists. As
any loading dose of a-2 agonists appears unwise, volume loading may be performed, then a very slow infusion may be commenced. Daily assessment of volemia is recommended.
Relative contra-indications:
Sick sinus syndrome.
Atrial fibrillation with a low ventricular response.
Atrioventricular block or a P+R interval of over 200 ms.
Prior treatment with beta-blockers resulting in a heart rate below 60 bpm, or prior administration of sympatholytic drugs.
Possible deleterious associations : diltiazem, urapidil.
Administration:
Avoid loading dose : use only continuous infusion over 24 h. Consider nasogastric route only for a limited period of time or
transdermal patch if peripheral perfusion is adequate and venous access or infusion syringes limited.
Clonidine : 0.4-2 mg.kg-1.h-1 [44]. Clonidine 1 mg.kg-1.h-1 without loading dose achieves 3%Ramsay%4 after 3-6 h (see
below : supplementation). If clonidine 2 mg.kg-1.h-1 is inadequate to achieve Ramsay&3 (e.g. delirium tremens) or escape
phenomenon after days or weeks, then supplement the a-2 agonist accordingly (see below).
Dexmedetomidine : 0.8-1.5 mg.kg-1.h-1 [6-7]. Despite the fact that dexmedetomidine is more selective for a-2 receptors, the
dosages collected throughout the literature do not reflect such a difference. A ceiling effect is reported above 1.5 mg.kg-1.h-1[45].

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

14

Journal of Intensive Care Medicine 000(00)


Supplemental sedation:

Rescue medication may be needed during 3-6 h i.e. progressive induction of sedation with slow infusion of a-2 agonists and
without loading dose : repeat midazolam 3-5 mg bolus, as needed, bearing in mind the respiratory depression induced by midazolam, e.g. in patients ventilated with pressure support-spontaneous ventilation.
Supplementation of a-2 agonists may be needed when high dose a-2 agonists are inadequate to get 3%Ramsay%4. Select drugs
which do
a)

not affect the respiratory drive (hydroxyzine i.v., loxapine through the nasogastric tube, ketamine, gamma-OH butyrate
[GammaOH1] . . . .). Such choice would be in line with a preserved spontaneous breathing e.g. under pressure support ventilation.
b) affect the respiratory drive : thiopentone, propofol, midazolam, opioid analgesics . . . Such choice implies to monitor closely
the respiratory status. Such combination does not allow one to assess which drug causes circulatory or cognitive disturbances:
a combination of e.g. clonidine-midazolam-sufentanyl-nicardipine is a door open to side effects and misunderstandings. In a
similar manner, adding propofol-remifentanil to clonidine may lead to profound hypotension-bradycardia.
If a clear cut indication exists, maintenance doses of hypnotic or halogenated agents are to be reduced by 80% or more and titrated
to sedation scale, e.g. 3%Ramsay%4 in patients with a tracheal tube.

Supplemental analgesia:
Consider pain medication only when clinically indicated, i.e. documented on pain scale.
Consider first drugs without effect on ventilatory drive : nefopam, low dose ketamine (alone or combined), when pressure
support-spontaneous ventilation is selected. A second choice would be opiates reduced by 80% or more, then titrated to pain scale.

Normothermia:
As a-2 agonists do not mask the respiratory consequences of hyperthermia (tachypnea) in septic patients, intentional
normothermia-mild hypothermia (36-37 ( C) may be warranted to lower respiratory rate. When hemodialysis is necessary, mild
intentional hypothermia is usually unnecessary. The effect of a-2 agonists on the hypothalamus makes the prescription of paracetamol unnecessary to prevent shivering.
Trouble shooting: bradycardia, hypotension, low cardiac output.
Reduce/eliminate halogenated, hypnotics, or other sedative agents.
Passive leg raising+Head-down tilt (beach chair position).
Volume loading upon induction and throughout a-2 agonist administration.
Vasoactive drugs: the requirement for ephedrine, phenylephrine, norepinephrine, isoproterenol, dobutamine are reduced.
As a-2 agonists are sympathetic inhibitors, a degree of hypotension is a possibility. After weighting the benefits of the a-2 agonist
vs. such a side effect, very low dose vasopressors5 may restore cerebral, coronary or renal perfusion pressure.
Military antishock trouser may be an option to reduce volume or vasopressor requirement.
For established bradycardia or low output, dobutamine requirements (titrated to echocardiographic assessment or superior vena
cava/mixed venous O2 saturation>70-75%) may be as low as 1.25 mg.kg-1.min-1.
For transient bradycardia, consider low-dose isoproterenol (titration to effect : boluses : 2-10 mg). If longer effect is needed,
atropine (0.75 to 1.2 mg).
Progressive tapering of clonidine over 2-4 days immediately after severing from non-invasive ventilation or prior to or following discharge from CCU : switching over from i.v. a-2 agonist to po clonidine is a simple option.

If considered, the authors recommend a slow i.v. infusion without bolus administration.
If considered, the authors recommend a slow i.v. infusion without bolus administration.

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

Pichot et al

15

References
1. Kollef MH, Levy NT, Ahrens TS, Schaiff R, Prentice D,
Sherman G. The use of continuous i.v. sedation is associated with
prolongation of mechanical ventilation. Chest. 1998;114(2):
541-548.
2. Kress JP, Pohlman AS, OConnor MF, Hall JB. Daily interruption
of sedative infusions in critically ill patients undergoing mechanical ventilation. N Engl J Med. 2000;342(20):1471-1477.
3. Knaus AE, Muthig V, Schickinger S, et al. Alpha2-adrenoceptor
subtypesunexpected functions for receptors and ligands
derived from gene-targeted mouse models. Neurochem Int.
2007;51(5):277-281.
4. Robertson D, Goldberg MR, Hollister AS, Wade D,
Robertson RM. Clonidine raises blood pressure in severe idiopathic orthostatic hypotension. Am J Med. 1983;74(2):193-200.
5. Shehabi Y, Ruettimann U, Adamson H, Innes R, Ickeringill M.
Dexmedetomidine infusion for more than 24 hours in critically ill
patients: sedative and cardiovascular effects. Intensive Care Med.
2004;30(12):2188-2196.
6. Riker RR, Shehabi Y, Bokesch PM, et al. Dexmedetomidine vs
midazolam for sedation of critically ill patients: a randomized
trial. J Am Med Assoc. 2009;301(5):489-499.
7. Pandharipande PP, Pun B, Herr DI, et al. Effect of sedation
with dexmedetomidine vs lorazepam on acute brain dysfunction
in mechanically ventilated patients: the MENDS randomized
controlled trial. J Am Med Assoc. 2007;298(22):2644-2653.
8. Marwaha J, Aghajanian GK. Relative potencies of alpha-1 and
alpha-2 antagonists in the locus coeruleus, dorsal raphe and dorsal
lateral geniculate nuclei: an electrophysiological study. J Pharmacol Exp Ther. 1982;222(2):287-293.
9. Quintin L, Gonon F, Buda M, Ghignone M, Hilaire G, Pujol JF.
Clonidine modulates locus coeruleus metabolic hyperactivity
induced by stress in behaving rats. Brain Res. 1986;362:
366-369.
10. Quintin L, Ghignone M. Central noradrenergic systems: basic and
clinical aspects relevant to anesthesia. In: Belmaker RH,
Sandler M, Dahlstrom A, eds. Progress in Catecholamine
Research. Part C: Clinical Aspects. (Proceedings of the Sixth
International Catecholamine Symposium). New York: A. R.
Liss; 1988:447-459.
11. Sun MK, Guyenet P. Effect of clonidine and gammaaminobutyric acid on the discharges of medullo-spinal sympathoexcitatory neurons in the rat. Brain Res. 1986;368(1):1-17.
12. Quintin L, Ghignone M. Risks associated with perioperative use
of alpha-2 adrenoceptors agonists. In: Scholz J, Tonner PH, eds.
Alpha-2 Adrenoceptor Agonists in Anaesthesia and Intensive
Care. London: Harcourt; 2000:347-368.
13. Toader E, Cividjian A, Rentero N, McAllen RM, Quintin L. Cardioinhibitory actions of clonidine assessed by cardiac vagal motoneuron recordings. J Hypertension. 2008;26:1169-1180.
14. Sleight P, West MJ, Korner PI, Oliver JR, Chalmers JP,
Robinson JL. The action of clonidine on the baroreflex control
of heart rate in conscious animals and man and on single aortic
baroreceptor discharge in the rabbit. Arch Int Pharmacodynamy
Ther. 1975;214:4-11.

15. Saunier CF, Akaoka H, de La Chapelle B, et al. Activation of


brain noradrenergic neurons during recovery from halothane
anesthesia: persistence of phasic activation after clonidine.
Anesthesiology. 1993;79(5):1072-1082.
16. Aston-Jones G, Ennis M, Pieribone VA, Nickell WT,
Shipley MT. The brain nucleus coeruleus: restricted afferent control of a broad efferent network. Science. 1986;234(4777):
734-737.
17. Segal IS, Vickery RG, Walton JK, Doze VA, Maze M. Dexmedetomidine diminishes halothane anesthetic requirements in rats
through a postsynaptic alpha-2 adrenergic receptor. Anesthesiology. 1988;69(6):818-823.
18. Kooner JS, Peart WS, Mathias CJ. The haemodynamic and hormonal responses after clonidine occur independently of sedation
in essential hypertension. Br J Clin Pharmacol. 1989;28(3):
249-255.
19. Schmitt H. The pharmacology of clonidine and related products.
In: Gross F, ed. Handbook of Experimental Pharmacology.
Berlin: Springer; 1977:299-396.
20. Hashimoto J, Chonan K, Aoki Y, et al. Therapeutic effects of evening administration of guanabenz and clonidine on morning
hypertension: evaluation using home-based blood pressure measurements. J Hypertens. 2003;21(4):805-811.
21. Onesti G, Bock KD, Heimsoth V, Kim KE, Merguet P. Clonidine: a new antihypertensive agent. Am J Cardiol. 1971;28(1):
74-83.
22. Wing LM, Reid JL, Davies DS, Dargie HJ, Dollery CT. Apparent
resistance to hypotensive effect of clonidine. Br Med J. 1977;
1(6054):136-138.
23. Miyazaki S, Uchida S, Mukai J, Nishihara K. Clonidine effects on
all-night human sleep: opposite action of low- and medium-dose
clonidine on human NREM-REM sleep proportion. Psychiatry
Clin Neurosci. 2004;58(2):138-144.
24. Huupponen E, Maksimow A, Lapinlampi P, et al. Electroencephalogram spindle activity during dexmedetomidine sedation and
physiological sleep. Acta Anaesthesiol Scand. 2008;52(2):
289-294.
25. Metz G, Nebel B. Clonidine in severe alcohol withdrawal delirium. Fortschr Med. 1983;101(26):1260-1264.
26. Spies CD, Dubisz N, Neumann T, et al. Therapy of alcohol withdrawal syndrome in intensive care unit patients following trauma:
results of a prospective, randomized trial. Crit Care Med. 1996;
24(3):414-422.
27. Bohrer H, Bach A, Layer M, Werming P. Clonidine as a sedative
adjunct in intensive care. Int Care Med. 1990;16:265-266.
28. Triltsch AE, Welte M, von HP, et al. Bispectral index-guided
sedation with dexmedetomidine in intensive care: a prospective,
randomized, double blind, placebo-controlled phase II study. Crit
Care Med. 2002;30(5):1007-1014.
29. Martin E, Ramsay G, Mantz J, Sum-Ping ST. The role of the
alpha-2 adrenoceptor agonist dexmedetomidine in postsurgical
sedation in the intensive care unit. J Intensive Care Med. 2003;
18(1):29-41.
30. Chrysostomou C, Di FS, Manrique AM, et al. Use of dexmedetomidine in children after cardiac and thoracic surgery. Pediatr Crit
Care Med. 2006;7(2):126-131.

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

16

Journal of Intensive Care Medicine 000(00)

31. Ruokonen E, Parviainen I, Jakob SM, et al. Dexmedetomidine


versus propofol/midazolam for long-term sedation during
mechanical ventilation. Intensive Care Med. 2009;35(2):282-290.
32. Gold MS, Redmond DE, Kleber HD. Clonidine blocks acute
opiate-withdrawal symptoms. Lancet. 1978;2(8090):599-602.
33. Liatsi D, Tsapas B, Pampori S, Tsagourias M, Pneumatikos I,
Matamis D. Respiratory, metabolic and hemodynamic effects of
clonidine in ventilated patients presenting with withdrawal syndrome. Intensive Care Med. 2009;35(2):275-281.
34. Freshwater-Turner D, Udy A, Lipman J, et al. Autonomic dysfunction in tetanuswhat lessons can be learnt with specific reference to alpha-2 agonists? Anaesthesia. 2007;62(10):1066-1070.
35. Sutton DN, Tremlett MR, Woodcock TE, Nielsen MS. Management
of autonomic dysfunction in severe tetanus: the use of magnesium
sulphate and clonidine. Intensive Care Med. 1990;16(2):75-80.
36. Martin J, Parsch A, Franck M, Wernecke KD, Fischer M, Spies C.
Practice of sedation and analgesia in German intensive care units:
results of a national survey. Crit Care. 2005;9(2):R117-R123.
37. Tonner PH, Weiler N, Paris A, Scholz J. Sedation and analgesia in
the intensive care unit. Curr Opin Anaesthesiol. 2003;16(2):
113-121.
38. Rubino AS, Onorati F, Caroleo S, et al. Impact of clonidine
administration on delirium and related respiratory weaning after
surgical correction of acute type-A aortic dissection: results of a
pilot study. Interact Cardiovasc Thorac Surg. 2010;10(1):58-62.
39. Maldonado JR, Wysong A, van der Starre PJ, Block T, Miller C,
Reitz BA. Dexmedetomidine and the reduction of postoperative
delirium after cardiac surgery. Psychosomatics. 2009;50(3):
206-217.
40. Venn RM, Bradshaw CJ, Spencer R, et al. Preliminary UK experience of dexmedetomidine, a novel agent for postoperative sedation in the intensive care unit. Anaesthesia. 1999;54(12):
1136-1142.
41. Venn RM, Grounds RM. Comparison between dexmedetomidine
and propofol for sedation in the intensive care unit: patient and
clinician perceptions. Br J Anaesth. 2001;87(5):684-690.
42. Venn RM, Hell J, Grounds RM. Respiratory effects of dexmedetomidine in the surgical patient requiring intensive care. Crit
Care. 2000;4(5):302-308.
43. Tobias JD. Dexmedetomidine: applications in pediatric critical
care and pediatric anesthesiology. Pediatr Crit Care Med. 2007;
2007:115-131.
44. Sauder P, Andreoletti M, Cambonie G, et al. Sedation and analgesia in intensive care. French Crit Care Soc. Ann Fr Anesth
Reanim. 2008;27:541-551.
45. Venn RM, Newman PJ, Grounds RM. A phase II study to evaluate
the efficacy of dexmedetomidine for sedation in the medical
intensive care unit. Intensive Care Med. 2003;29:201-207.
46. MacLaren R, Forrest LK, Kiser TH. Adjunctive dexmedetomidine therapy in the intensive care unit: a retrospective assessment
of impact on sedative and analgesic requirements, levels of sedation and analgesia, and ventilatory and hemodynamic parameters.
Pharmacotherapy. 2007;27(3):351-359.
47. Connor DF, Fletcher KE, Swanson JM. A meta-analysis of
clonidine for symptoms of attention-deficit hyperactivity disorder.
J Am Acad Child Adolesc Psychiatry. 1999;38(12):1551-1559.

48. Cohen DJ, Detlor J, Young JG, Shaywitz BA. Clonidine ameliorates Gilles de la Tourette syndrome. Arch Gen Psychiatry. 1980;
37(12):1350-1357.
49. Freedman R, Kirch D, Bell J, et al. Clonidine treatment of schizophrenia. Double-blind comparison to placebo and neuroleptic
drugs. Acta Psychiatry Scand. 1982;65:35-45.
50. McEntee WJ, Mair RG. Memory enhancement in Korsakoffs
psychosis by clonidine: further evidence for a noradrenergic deficit. Ann Neurol. 1980;7:466-470.
51. Akada S, Takeda S, Yoshida Y, et al. The efficacy of dexmedetomidine in patients with noninvasive ventilation: a preliminary
study. Anesth Analg. 2008;107(1):167-170.
52. Payen D, Quintin L, Plaisance P, Chiron B, Lhoste F. Head injury:
clonidine decreases plasma catecholamines. Crit Care Med. 1990;
18(4):392-395.
53. Kariya N, Shindoh M, Nishi S, Yukioka H, Asada A. Oral clonidine to control hypertension after head injury. Eur J Emerg Med.
1999;6(1):77-79.
54. Ter Minassian A, Beydon L, Decq P, Bonnet F. Changes in cerebral hemodynamics after a single dose of clonidine in severely
head-injured patients. Anesth Analg. 1997;84(1):127-132.
55. Aryan HE, Box KW, Ibrahim D, Desiraju U, Ames CP. Safety and
efficacy of dexmedetomidine in neurosurgical patients. Brain Inj.
2006;20(8):791-798.
56. Asgeirsson B, Grande PO, Nordstrom CH. A new therapy of
post-trauma brain oedema based on haemodynamic principles
for brain volume regulation. Intensive Care Med. 1994;20(4):
260-267.
57. Eker C, Asgeirsson B, Grande PO, Schalen W, Nordstrom CH.
Improved outcome after severe head injury with a new therapybased on principles for brain volume regulation and preserved microcirculation. Crit Care Med. 1998;26(11):
1881-1886.
58. Naredi S, Eden E, Zall S, Stephensen H, Rydenhag B. A standardized neurosurgical neurointensive therapy directed toward vasogenic edema after severe traumatic brain injury: clinical results.
Intensive Care Med. 1998;24(5):446-451.
59. Grande PO, Asgeirsson B, Nordstrom CH. Volume-targeted
therapy of increased intracranial pressure: the Lund concept
unifies surgical and non-surgical treatments. Acta Anaesthesiol
Scand. 2002;46(8):929-941.
60. Paalzow G, Paalzow L. Clonidine antinociceptive activity: effects
of drugs influencing central monoaminergic and cholinergic
mechanisms in the rat. Naunyn Schmiedebergs Arch Pharmacol.
1976;292(2):119-126.
61. Skingle M, Hayes AG, Tyers MB. Antinociceptive activity of
clonidine in the mouse, rat and dog. Life Sci. 1982;31(11):
1123-1132.
62. Marwaha J, Kehne JH, Commisaris RL, Lakovski J, Shaw W,
Davis M. Spinal clonidine inhibits neural firing in locus coeruleus. Brain Res. 1983;276(2):379-382.
63. Kauppila T, Kemppainen P, Tanila H, Pertovaara A. Effect of systemic medetomidine, an alpha-2 adrenoceptor agonist, on experimental pain in humans. Anesthesiology. 1991;74(1):3-8.
64. Ghignone M, Quintin L, Duke PC, Kehler CH, Calvillo O. Effects
of clonidine on narcotic requirements and hemodynamic response

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

Pichot et al

65.

66.

67.

68.
69.

70.

71.

72.

73.

74.

75.

76.
77.

78.

79.

80.
81.

17

during induction of fentanyl anesthesia and endotracheal intubation. Anesthesiology. 1986;64(1):36-42.


Ghignone M, Calvillo O, Quintin L. Anesthesia and hypertension:
the effect of clonidine on perioperative hemodynamics and
isoflurane requirements. Anesthesiology. 1987;67(1):3-10.
Ghignone M, Noe C, Calvillo O, Quintin L. Anesthesia for
ophthalmic surgery in the elderly: the effects of clonidine on
intraocular pressure, perioperative hemodynamics and anesthetic
requirements. Anesthesiology. 1988;68(5):707-716.
De Kock M, Wiederkehr P, Laghmiche A, Scholtes J-L. Epidural
clonidine used as the sole analgesic agent during and after
abdominal surgery. Anesthesiology. 1997;86(2):285-292.
Ramsay MA, Luterman DL. Dexmedetomidine as a total intravenous anesthetic agent. Anesthesiology. 2004;101(3):787-790.
Hofer RE, Sprung J, Sarr MG, Wedel DJ. Anesthesia for a patient
with morbid obesity using dexmedetomidine without narcotics.
Can J Anaesth. 2005;52(2):176-180.
Ramsay MA, Saha D, Hebeler RF. Tracheal resection in the morbidly obese patient: the role of dexmedetomidine. J Clin Anesth.
2006;18(6):452-454.
Voituron N, Kinkead R, Quintin L, Hilaire G. Clonidine pretreatment alleviates the depressive respiratory effects of benzodiazepine in en bloc medullary preparations from neonatal
mice. International Symposium on Respiratory Control.
Saint-Maximin: France; 2008.
Bolme P, Fuxe K. Pharmacological studies on a possible role of
central noradrenaline neurons in respiratory control. J Pharm
Pharmacol. 1973;25(4):351-352.
Bolme P, Corrodi H, Fuxe K, Hokfelt T, Lidbrink P, Goldstein M.
Possible involvement of central adrenaline neurons in vasomotor
and respiratory control. Studies with clonidine and its interactions
with piperoxane and yohimbine. Eur J Pharmacol. 1974;28(1):
89-94.
Furst SR, Weinger MB. Dexmedetomidine, a selective alpha 2agonist, does not potentiate the cardiorespiratory depression of
alfentanil in the rat. Anesthesiology. 1990;72(5):882-888.
Bailey PL, Speng RJ, Johnson GK, et al. Respiratory effects of
clonidine alone and combined with morphine in humans.
Anesthesiology. 1991;74(1):43-48.
Ooi R, Pattison J, Feldman SA. The effects of intravenous clonidine on ventilation. Anaesthesia. 1991;46(8):632-633.
Takahashi H, Nishikawa T, Mizutani T, Handa F. Oral clonidine
premedication decreases energy expenditure in human volunteers. Can J Anaesth. 1997;44(3):268-272.
Quintin L, Viale JP, Annat G, et al. Oxygen uptake after major
abdominal surgery: effect of clonidine. Anesthesiology. 1991;
74(2):236-241.
Pawlik MT, Hansen E, Waldhauser D, Selig C, Kuehnel TS. Clonidine premedication in patients with sleep apnea syndrome: a
randomized, double-blind, placebo-controlled study. Anesth
Analg. 2005;101(5):1374-1380.
Bhatt J, Primhak, R, Mayer, A. Clonidine and noninvasive ventilation in children. Eur Respir Soc. 2005:P638.
Lindgren BR, Ekstrom T, Andersson RGG. The effects of inhaled
clonidine in patients with asthma. Am Rev Resp Dis. 1986;134:
266-269.

82. Dinh Xuan AT, Lockhart A. Bronchial effects of alpha-2 adrenoceptor agonists and of other antihypertensive agents in asthma.
Am J Med. 1989;87:34S-37S.
83. Groeben H, Mitzner W, Brown RH. Effects of the alpha-2 adrenoceptor agonist dexmedetomidine on bronchoconstriction in
dogs. Anesthesiology. 2004;100:359-363.
84. Tobias JD, Berkenbosch JW, Russo P. Additional experience
with dexmedetomidine in pediatric patients. South Med J. 2003;
96(9):871-875.
85. Arpino PA, Kalafatas K, Thompson BT. Feasibility of dexmedetomidine in facilitating extubation in the intensive care unit.
JClin Pharm Ther. 2008;33(1):25-30.
86. Kobinger W, Pichler L. The central modulatory effect of
clonidine on the cardiodepressor reflex after suppression of
synthesis and storage of noradrenaline. Eur J Pharmacol. 1975;
30(1):56-62.
87. Haeusler G. Clonidine-induced inhibition of sympathetic nerveactivity: no indication for a central presynaptic or an indirect sympathomimetic mode of action. Naunyn. 1974;286:
97-111.
88. Mermet C, Quintin L. Effect of clonidine on the catechol metabolism in the rostral ventrolateral medulla: an in vivo electrochemical study. Eur J Pharmacol. 1991;204(1):105-107.
89. Guyenet P. Is the hypotensive effect of clonidine and related
drugs due to imidazoline binding sites? Am J Physiol. 1997;
273(5 Pt 2):R1580-R1584.
90. Bousquet P, Feldman J, Schwartz J. Central cardiovascular
effects of alpha adrenergic drugs: differences between catecholamines and imidazolines. J Pharmacol Exp Ther. 1984;
230(1):232-236.
91. Gillon JY, Quintin L, Ghignone M, Pujol JF. Clonidine modulates
the ventrolateral medullary catechol metabolic hyperactivity
induced by hypotension. Brain Res. 1987;418(1):157-163.
92. Heusch G, Schipke J, Thamer V. Clonidine prevents the sympathetic initiation and aggravation of poststenotic myocardial ischemia. J Cardiovasc Pharmacol. 1985;7(6):1176-1182.
93. Lawrence CJ, Prinzen FW, de LS. The effect of dexmedetomidine
on the balance of myocardial energy requirement and oxygen supply
and demand. Anesth Analg. 1996;82(3):544-550.
94. Motz W, Ippisch R, Strauer BE. The role of clonidine in hypertensive heart disease. Influence on myocardial contractility and
left ventricular afterload. Chest. 1983;83(suppl 2):433-435.
95. Giles TD, Iteld BJ, Mautner RK, Rognoni PA, Dillenkoffer RL.
Short-term effects of intravenous clonidine in congestive heart
failure. Clin Pharmacol Ther. 1981;30(6):724-728.
96. Hermiller JB, Magorien RD, Leithe ME, Unverferth DV,
Leier CV. Clonidine in congestive heart failure: a vasodilator
with negative inotropic effects. Am J Cardiol. 1983;51(5):
791-795.
97. Manolis AJ, Olympios C, Sifaki M, et al. Suppressing sympathetic activation in congestive heart failure: a new therapeutic
strategy. Hypertension. 1995;26(5):719-724.
98. Olivari MT, Levine TB, Cohn JN. Acute hemodynamic and hormonal effects of central vs. peripheral sympathetic inhibition in
patients with congestive heart failure. J Cardiovasc Pharmacol.
1986;8(5):973-978.

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

18

Journal of Intensive Care Medicine 000(00)

99. Flacke JW, Flacke WE. Clonidine prevention of myocardial


ischemia during cardiac surgery: will this change outcome ? J
Cardiothorac Vasc Anesth. 1993;7(4):383-385.
100. Giles TD, Thomas MG, Quiroz A, Rice JC, Plauche W,
Sander GE. Acute and short-term effects of clonidine in heart
failure. Angiology. 1987;38(7):537-548.
101. Foresti A, Massari FM, Lotto A. Hemodynamic effects of clonidine
in patients with acute myocardial infarction complicated by hypertension. J Cardiovasc Pharmacol. 1986;8(suppl 3):S30-S32.
102. Mitrovic V, Hallier E, Kuschke HJ. The hemodynamic effects of
clonidine in patients with grade III to IV hypertension. J Cardiovasc Pharmacol. 1986;8(suppl 3):S51-S55.
103. Northam WJ, Mobley P. Clonidine pretreatment enhances the
sensitivity of the beta-noradrenergic receptor coupled adenylate
cyclase system in astrocytes. Eur J Pharmacol. 1985;113(1):
153-154.
104. Zoukos Y, Thomaides T, Pavitt DV, Leonard JP, Cuzner ML,
Mathias CJ. Upregulation of beta adrenoceptors on circulating
mononuclear cells after reduction of central sympathetic outflow
by clonidine in normal subjects. Clin Auton Res. 1992;2:
165-170.
105. Quiroz AC, Eilen SD, Sander GE, Giles TD. The effect of
intravenous clonidine hydrochloride on the isolated forearm
venous segment in heart failure. Chest. 1983;83(suppl 2):
430-433.
106. Guthrie GP, Kotchen TA. Effects of oral clonidine on baroreflex
function in patients with essential hypertension. Chest. 1983;
83(suppl 2):327-328.
107. Dollery CT, Harington M, Hodge JV. Haemodynamic studies
with methyldopa: effect on cardiac output and response to
pressor amines. Br Heart J. 1963;25:670-676.
108. Parlow J, Sagnard P, Begou G, Viale JP, Quintin L. The effects
of clonidine on sensitivity to phenylephrine and nitroprusside in
patients with essential hypertension recovering from surgery.
Anesth Analg. 1999;88(6):1239-1243.
109. Quintin L, Bouilloc X, Butin E, et al. Clonidine for aortic surgery in hypertensive patients: a double blind controlled randomized study. Anesth Analg. 1996;83:687-695.
110. De Kock M, Laterre PF, Van Obbergh L, Carlier M, Lerut J. The
effects of intraoperative intravenous clonidine on fluid requirements, hemodynamic variables, and support during liver transplantation: a prospective, randomized study. Anesth Analg.
1998;86(3):468-476.
111. Herr DL, Sum-Ping ST, England M. ICU sedation after coronary
artery bypass graft surgery: dexmedetomidine-based versus
propofol-based sedation regimens. J Cardiothorac Vasc Anesth.
2003;17(5):576-584.
112. Pandharipande PP, Sanders RD, Girard TD, et al. Effect of dexmedetomidine versus lorazepam on outcome in patients with
sepsis: an a priori-designed analysis of the MENDS randomized
controlled trial. Crit Care. 2010;14(2):R38.
113. Dodd-o JM, Breslow MJ, Dorman T, Rosenfeld BA. Preserved
sympathetic response to hypotension despite perioperative alpha2 agonist administration. Anesth Analg. 1997;84(6):1208-1210.
114. Pichot C, Mathern P, Khettab F, Ghignone M, Geloen A, Quintin
L. Increased pressor response to noradrenaline during septic

115.
116.

117.

118.
119.

120.

121.

122.

123.

124.

125.
126.

127.

128.
129.

130.

131.

132.

shock following clonidine? Anesthesia Intensive Care (Sydney).


2010;38, 734-735.
Tobias JD. Bradycardia during dexmedetomidine and therapeutic hypothermia. J Intensive Care Med. 2008;23:403-408.
Pohl-Schickinger A, Lemmer J, Hubler M, et al. Intravenous clonidine infusion in infants after cardiovascular surgery. Paediatr
Anaesth. 2008;18(3):217-222.
Arenas-Lopez S, Riphagen S, Tibby SM, et al. Use of oral clonidine for sedation in ventilated paediatric intensive care
patients. Intensive Care Med. 2004;30(8):1625-1629.
Czaja AS, Zimmerman JJ. The use of dexmedetomidine in critically ill children. Pediatr Crit Care Med. 2009;10(3):381-386.
Gerlach AT, Dasta JF, Steinberg S, Martin LC, Cook CH. A new
dosing protocol reduces dexmedetomidine-associated hypotension in critically ill surgical patients. J Crit Care. 2009;24(4):
568-574.
Jorden VS, Pousman RM, Sanford MM, Thorborg PA,
Hutchens MP. Dexmedetomidine overdose in the perioperative
setting. Ann Pharmacother. 2004;38(5):803-807.
Meyer C, Cambray R. One hundred times the intended dose of
caudal clonidine in three pediatric patients. Paediatr Anaesth.
2008;18(9):888-890.
Erkonen G, Lamb F, Tobias JD. High-dose dexmedetomidineinduced hypertension in a child with traumatic brain injury. Neurocrit Care. 2008;9(3):366-369.
Harron DW, Kobinger W. Facilitation of the Bezold-Jarisch
reflex by central stimulation of alpha-2 adrenoceptors in dogs.
Naunyn Schmiedebergs Arch Pharmacol. 1984;325(3):193-197.
Clementy J, Castagne D, Wicker P, Dallochio M, Bricaud H.
Study of the electrophysiologic properties of clonidine administered intravenously. J Cardiovasc Pharmacol. 1986;8(suppl 8):
S24-S29.
Schwartz E, Friedman E, Mouallem M, Farfel Z. Sinus arrest associated with clonidine therapy. Clin Cardiol. 1988;11(1):53-54.
Ingersoll-Weng E, Manecke GR Jr, Thistlethwaite PA. Dexmedetomidine and cardiac arrest. Anesthesiology. 2004;100(3):
738-739.
Roulot D, Moreau R, Gaudin C, et al. Long-term sympathetic
and hemodynamic responses to clonidine in patients with cirrhosis and ascites. Gastroenterology. 1992;102(4 Pt 1):1309-1318.
Arndts D. New aspects of the clinical pharmacology of clonidine. Chest. 1983;83(suppl 2):397-400.
Lenaerts A, Codden T, Meunier JC, Henry JP, Ligny G. Effects
of clonidine on diuretic response in ascitic patients with cirrhosis
and activation of sympathetic nervous system. Hepatology.
2006;44(4):844-849.
Esler M, Dudley F, Jennings G, et al. Increased sympathetic nervous activity and the effects of its inhibition with clonidine in
alcoholic cirrhosis. Ann Intern Med. 1992;116(6):446-455.
Rubinoff MJ, Piccione PR, Holt PR. Clonidine prolongs human
small intestine transit time: use of the lactulose-breath hydrogen
test. Am J Gastroenterol. 1989;84(4):372-374.
Morali GA, Braverman DZ, Lissi J, Goldstein R, Jacobsohn WZ.
Effect of clonidine on gallbladder contraction and small bowel
transit time in insulin-treated diabetics. Am J Gastroenterol.
1991;86(8):995-999.

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

Pichot et al

19

133. Bear R, Steer K. Pseudo-obstruction due to clonidine. Br Med J.


1976;1(6003):197.
134. Stieger DS, Cantieni R, Frutiger A. Acute colonic pseudoobstruction (Ogilvies syndrome) in two patients receiving high
dose clonidine for delirium tremens. Intensive Care Med.
1997;23(7):780-782.
135. Hamaya Y, Nishikawa T, Dohi S. Diuretic effect of clonidine
during isoflurane-nitrous oxide and oxygen anesthesia. Anesthesiology. 1994;81(4):811-819.
136. Liepert DJ, Townsend GE. Improved hemodynamic and renal
function with clonidine in coronary artery bypass grafting.
Anesth Analg. 1990;70:S240.
137. Kulka PJ, Tryba M, Zenz M. Preoperative alpha-2 adrenergic
receptor agonists prevent the deterioration of renal function after
cardiac surgery: results of a randomized, controlled trial. Crit
Care Med. 1996;24:947-952.
138. De Wolf AM, Fragen RJ, Avram MJ, Fitzgerald PC, RahimiDanesh F. The pharmacokinetics of dexmedetomidine in volunteers with severe renal impairment. Anesth Analg. 2001;93(5):
1205-1209.
139. Hoffmann BB, Lefkowitz RJ. Catecholamines, sympathomimetic
drugs and adrenergic receptor antagonists. In: Hardman JG,
Limbird LE, eds. Goodmans and Gilmans The pharmacological
Basis of Therapeutics. New York: McGraw-Hill; 1996:199-248.
140. Venn RM, Karol MD, Grounds RM. Pharmacokinetics of
dexmedetomidine infusions for sedation of postoperative
patients requiring intensive care. Br J Anaesth. 2002;88(5):
669-675.
141. Mertes N, Goeters C, Kuhmann M, Zander JF. Postoperative
alpha-2 adrenergic stimulation attenuates protein catabolism.
Anesth Analg. 1996;82(2):258-263.
142. Kulka PJ, Tryba M, Reimer T, Weisser H. Clonidine prevents
tissue-malperfusion during extracorporal circulation. Anesth
Analg. 1996;83:482-487.
143. von Dossow V, Baehr N, Moshirzadeh M, et al. Clonidine
attenuated early proinflammatory response in T-cell subsets
after cardiac surgery. Anesth Analg. 2006;103(4):809-814.
144. Hofer S, Steppan J, Wagner T, et al. Central sympatholytics prolong survival in experimental sepsis. Crit Care. 2009;13(1):R11.
145. Taniguchi T, Kidani Y, Kanakura H, Takemoto Y, Yamamoto K.
Effects of dexmedetomidine on mortality rate and inflammatory

146.

147.

148.
149.

150.

151.

152.

153.

154.

155.
156.

157.

response to endotoxin-induced shock in rats. Crit Care Med.


2004;32(6):1322-1326.
Taniguchi T, Kurita A, Kobayashi K, Yamamoto K, Inaba H.
Dose- and time-related effects of dexmedetomidine on mortality
and inflammatory responses to endotoxin-induced shock in rats.
J Anesth. 2008;22(3):221-228.
Venn RM, Bryant A, Hall GM, Grounds RM. Effects of dexmedetomidine on adrenocortical function, and the cardiovascular,
endocrine and inflammatory responses in post-operative patients
needing sedation in the intensive care unit. Br J Anaesth. 2001;
86(5):650-656.
Wunsch H, Kress JP. A new era for sedation in ICU patients. J
Am Med Assoc. 2009;301(5):542-544.
Esmaoglu A, Ulgey A, Akin A, Boyaci A. Comparison between
dexmedetomidine and midazolam for sedation of eclampsia
patients in the intensive care unit. J Crit Care. 2009;24(4):
551-555.
Oliver MF, Goldman L, Julian DG, Holme I. Effect of mivazerol
on perioperative cardiac complications during non-cardiac surgery in patients with coronary disease. Anesthesiology. 1999;
91(4):951-961.
Wallace AW, Galindez D, Salalieh A, et al. Effect of clonidine
on cardiovascular morbidity and mortality after noncardiac surgery. Anesthesiology. 2004;101(2):284-293.
Wijeysundera DN, Naik JS, Beattie S. Alpha-2 adrenergic agonists to prevent cardiovascular complications. Am J Med. 2003;
114:752.
Stevens RD, Burri H, Tramer MR. Pharmacologic myocardial
protection in patients undergoing noncardiac surgery: a quantitative systematic review. Anesth Analg. 2003;97(3):623-633.
Gregorakos L, Kerezoudi E, Dimopoulos G, Thomaides T. Management of blood pressure instability in severe tetanus: the use
of clonidine. Intensive Care Med. 1997;23(8):893-895.
Tracey KJ. The inflammatory reflex. Nature. 2002;420(6917):
853-859.
Miles BA, Lafuse WP, Zwilling BS. Binding of alphaadrenergic receptors stimulates the anti-mycobacterial activity
of murine peritoneal macrophages. J Neuroimmunol. 1996;
71(1-2):19-24.
Shelly MP. Dexmedetomidine: a real innovation or more of the
same? Br J Anaesth. 2001;87:677-678.

Downloaded from jic.sagepub.com at CAPES on April 29, 2011

S-ar putea să vă placă și