Anda di halaman 1dari 12

REVIEW ARTICLE

Neuroprotection by Ketamine: A Review of the Experimental and Clinical Evidence


Judith A. Hudetz, PhD, and Paul S. Pagel, MD, PhD

N EUROPROTECTION MAY BE DEFINED AS the “pre-


vention or amelioration of neuronal damage evidenced by
abnormalities in cerebral metabolism, histopathology or neuro-
that rapidly lead to cell death, including excitotoxicity, peri-
infarct depolarization, inflammation, and apoptosis.3 Focal im-
pairment of CBF restricts the delivery of O2 and glucose,
logic function occurring after a hypoxic or an ischemic event.”1 thereby impairing the generation of energy required to maintain
Thus, the prevention of cerebral ischemia and the recovery of ionic gradients.4 As a result, intracellular energy supplies are
neural tissue that already has sustained an ischemic insult rapidly exhausted, membrane potential is lost, and neurons and
represent essential goals of neuroprotection. Neuroprotection their supporting glial cells depolarize.5 Somatodendritic and
may occur as a consequence of reduced O2 demand, enhanced presynaptic voltage-dependent Ca2⫹ channels become activated
O2 delivery, or attenuation of pathologic processes that con- under these conditions, and excitatory amino acids (eg, gluta-
tribute to cellular injury or death. For decades, the dissociative mate) are released into the extracellular space. Energy-depen-
sedative-hypnotic ketamine has been considered to be rela- dent homeostatic processes, such as presynaptic reuptake of
tively contraindicated in the presence of ischemic brain injury excitatory amino acids are simultaneously impeded, thereby
or an intracerebral mass because the drug increases cerebral further increasing the accumulation of glutamate in the extra-
metabolic rate for O2 (CMRO2), cerebral blood flow (CBF), cellular space. The NMDA-receptor controls an ion channel
and intracranial pressure (ICP).2 However, this traditionally that is permeable to Ca2⫹ and glutamate release by ischemic
held supposition may not be entirely correct because ketamine neurons. The activation of these receptors increases Ca2⫹ influx
also may have the potential to exert important neuroprotective and rapidly initiates cell necrosis and apoptosis.6 Cerebral
effects against ischemic damage. Ketamine inhibits N-methyl- neurons that are subjected to ischemia-induced glutamate-re-
D-aspartate (NMDA) receptor activation and excitotoxic sig- ceptor activation, Ca2⫹ overload, O2-derived free radicals, or
naling, reduces neuronal apoptosis (programmed cell death),
mitochondrial and DNA damage may be irreversibly damaged
attenuates the systemic inflammatory response to tissue injury,
depending on the source and intensity of the injury, the type of
and also maintains cerebral perfusion pressure as a result of
neural cell, and its developmental stage.7 Thus, a potential
sympathetic nervous system activation. These actions of ket-
therapeutic approach to prevent this sequence of events is the
amine may act to offset its detrimental effect on cerebral blood
blockade of NMDA receptors.8 As a result, ketamine may
flow and metabolism, especially under conditions in which
potentially inhibit neuronal cell death by preventing excitotoxic
arterial CO2 tension is controlled during mechanical ventilation. In
injury through this mechanism.9
the setting of cardiac surgery with or without cardiopulmonary
The intracellular signaling pathways activated during exci-
bypass (CPB), cerebral ischemia may be characterized by global
or regional dysfunction resulting from hypoperfusion and micro- totoxicity trigger the expression of genes that initiate inflam-
embolization. mation, another important contributor to ischemic injury. The
The authors review the current evidence supporting the hy- Ca2⫹-induced activation of intracellular second messengers (eg,
pothesis that ketamine may protect the human brain from ceramide), the release of O2-derived free radicals, and the
ischemic injury. A search of the medical literature was con- presence of hypoxia itself stimulate transcription and transla-
ducted by using several bibliographic databases (PubMed, tion of nuclear factor-␬B and hypoxia-inducible factor 1.10,11
CINAHL, Cochrane, and PsychInfo) using the following medical Ketamine suppresses nuclear factor-␬B expression12 involved
subject headings: “ketamine,” “neuroprotection,” “neurologic,”
“stroke,” “ischemia,” “clinical,” and “human.” Forty-four arti-
From the Department of Anesthesiology, Medical College of Wis-
cles were identified, 11 of them in languages other than En-
consin and Clement J. Zablocki Veterans Administration Medical Cen-
glish. The review of the 33 articles and the abstracts of the ter, Milwaukee, WI.
non-English articles were supplemented by a manual search of Supported in part by the Medical College of Wisconsin Institutional
related articles. Grant RAC 508-3 and by departmental funds.
Address reprint requests to Judith A. Hudetz, PhD, Department of
O2 DEPRIVATION AND MECHANISMS Anesthesiology, Clement J. Zablocki Veterans Administration Medical
OF EXCITOTOXICITY Center, 5000 W National Avenue, Milwaukee, WI 53295. E-mail: judith.
hudetz@va.gov
Cerebral neurons rapidly consume O2 and glucose and de- © 2010 Elsevier Inc. All rights reserved.
pend almost exclusively on these substrates to drive oxidative 1053-0770/10/2401-0025$36.00/0
phosphorylation for energy production and are extremely sen- doi:10.1053/j.jvca.2009.05.008
sitive to reductions in the O2 and glucose delivery.3 Restriction Key words: ketamine, neuroprotection, neurologic, stroke, ischemia,
of O2 and glucose delivery to the brain causes a series of events clinical, human

Journal of Cardiothoracic and Vascular Anesthesia, Vol 24, No 1 (February), 2010: pp 131-142 131
132 HUDETZ AND PAGEL

in the transcription of genes encoding the proinflammatory ministration of racemic ketamine before the initiation of hypo-
cytokines tumor necrosis factor ␣ (TNF-␣), interleukin (IL)-6, capnia reduced white-matter injury, neuronal damage, and as-
and IL-8.13 Although microvascular obstruction by neutrophils troglial proliferation. These data suggested that racemic
may worsen the degree of ischemia,14-16 the production of toxic ketamine may be beneficial for preventing brain dysfunction
mediators (eg, TNF-␣) by these activated inflammatory cells caused by inadvertent hypocapnia during mechanical ventila-
and injured neurons also contributes to further exacerbation of tion. Racemic ketamine also enhanced neurologic outcome
ischemic injury during early reperfusion. For example, isch- concomitant with reductions in plasma catecholamine concen-
emic neurons produce the proinflammatory cytokine TNF-␣ trations in a rat model of incomplete cerebral ischemia.28 The
that has been linked to accelerated ischemic injury through a beneficial effects of ketamine on cerebral histopathology were
“feed-forward” mechanism.17 Thus, ketamine administration dose-dependent. Ketamine (20 mg/kg) did not affect neuronal
may affect this process by reducing brain injury during early damage in the selectively vulnerable hippocampal CA1 region
reperfusion. in a rat model of near-complete forebrain ischemia, but larger
doses of ketamine administered during the reperfusion pro-
NEUROPROTECTIVE EFFECTS OF KETAMINE: vided significant neuroprotection.29 These results suggested
EXPERIMENTAL EVIDENCE that NMDA receptor–mediated events may contribute to neu-
Several experimental studies in vitro and in vivo suggested ronal damage in selectively vulnerable regions of the central
that ketamine may be capable of producing neuroprotective nervous system after the ischemia insult and further implied
effects.9,18-20 Glutamatergic signaling has been linked to nitric that ketamine may attenuate such damage. Despite the intrigu-
oxide metabolism.21 Racemic ketamine inhibited nitric oxide ing nature of these findings, it is important to emphasize that
synthesis and nitric oxide-dependent cyclic guanosine mono- animal models of cerebral ischemia and their modulation by
phosphate production stimulated by glutamate and glutamate NMDA-receptor antagonists may not predict results in humans.
analogs (eg, NMDA, quisqualate, and kainite) in primary cul- In addition, the wide variation in effective doses of ketamine
tures of cortical neurons and glia.19 These findings suggested reported within and between animal species suggests that the
that this ketamine-induced inhibition of glutamate receptors dose of ketamine required for possible neuroprotection in hu-
may contribute not only to anesthesia but also cellular protec- mans is not immediately apparent.
tion against ischemic injury.
Racemic and (S)- but not (R)-ketamine attenuated injury CEREBROVASCULAR AND METABOLIC EFFECTS OF
after glutamate exposure or axonal transection in rat hippocam- KETAMINE: EXPERIMENTAL FINDINGS
pal neurons in vitro.9 Neuroregenerative effects also were ob- Low doses of ketamine (⬍2 mg/kg) have been reported to
served with (S)-ketamine but not racemic or (R)-ketamine.9 increase30-33 but also not to affect CBF and CMRO2 in exper-
Synthesis of a growth-associated protein related to plasticity imental animals. These apparently conflicting observations
and repair were enhanced after glutamate injury by (S)- but not may be explained at least in part because ketamine may inhibit
racemic ketamine in rat hippocampal neurons.18 To determine metabolism in selected brain regions while simultaneously en-
the relative neuroprotective activity of (S)-ketamine compared hancing metabolism in others concomitant with changes in
with its (R)-isomer administered after the ischemic injury in CBF.30,34,35 Racemic and (S)-ketamine decreased glucose cere-
vivo, the functional and neurohistologic outcomes of rats ex- bral metabolic rate (CMRglu) in cortical, thalamic, cerebellar,
posed to global forebrain ischemia were examined.22 (S)- but and brainstem regions but increased CMRglu in limbic areas. In
not (R)-ketamine significantly reduced neuronal cell loss in the contrast, (R)-ketamine caused less-pronounced reductions in
cerebral cortex. (S)- but not (R)-ketamine also restored the CMRglu in 11 brain areas, whereas this optical isomer mark-
cortical O2 saturation to preischemic values independent of edly increased CMRglu in the basal ganglia and limbic regions.
CBF. Thus, the (S)-ketamine may reduce cortical neuronal This regional specificity of ketamine on CMRglu and CBF was
damage after cerebral ischemia by improving the ratio of O2 consistent with the heterogenous effects of the drug on brain
supply to consumption during reperfusion.22 (S)-ketamine also electrical activity.36-38 The actions of ketamine on regional
favorably influenced neuronal apoptosis by attenuating in- cerebral perfusion and metabolism appear to be sensitive to
creases in expression of the proapoptotic Bax protein produced preexisting cerebrovascular tone and the prevalence of other
by cerebral ischemia and reperfusion in rats compared with anesthetics.39-44 The balance between microregional O2 demand
those that did not receive the drug.23 These data emphasize that and supply has been shown to be heterogenously distributed.45
the salutary actions of ketamine in models of neuronal injury The influence of ketamine on regional CBF and microregional
appear to be stereoselective. arterial and venous O2 saturation were determined in the ante-
Ketamine also may decrease the severity of cerebral injury rior cortex, posterior cortex, and pons in rats. In this setting,
by interfering with the inflammatory response to ischemia. ketamine did not substantially affect regional CBF or O2 sup-
Ketamine suppressed lipopolysaccharide-induced TNF-␣, IL-6, ply-demand relations in contrast to enhanced CBF and CMRO2
and IL-8 production and inhibited neutrophil adhesion to the typically reported.45
endothelium in vitro.24,25 Racemic ketamine improved the neu- Nimkoff et al39 examined the effects of intravenous anes-
rologic severity score at 24 and 48 hours and reduced the thetics on ICP and cerebral perfusion pressure (CPP) in 2
volume of hemorrhagic necrosis without altering cerebral models of brain edema in cats. A space-occupying lesion was
edema after head trauma in rats.26 Ketamine also attenuated used to mimic vasogenic edema, whereas cytotoxic brain
neuronal damage in the caudoputamen of rats exposed to edema was created by an acute reduction in blood osmolality.
chronic, hypocapnia-induced cerebral hypoperfusion.27 The ad- Ketamine reduced ICP caused by space-occupying lesions to a
NEUROPROTECTION BY KETAMINE 133

similar degree as methohexital and propofol, but the dissocia- ways that extensively innervate the cerebral cortex may play an
tive anesthetic also increased CPP, in contrast to the other important role in this phenomenon. Whether the NMDA an-
drugs. However, ketamine, methohexital, and propofol did not tagonist-induced neurodegeneration observed in rats also oc-
affect ICP or CPP in the cytotoxic brain edema model. These curs in humans is presently unknown. Notably, the doses and
data suggested that ketamine may attenuate the increase in ICP durations of administration of ketamine-implicated neurotoxic-
caused by space-occupying lesions but may be detrimental ity in experimental animals often do not correlate with and
during generalized edema. The authors postulated that some frequently greatly exceed those used to produce sedation or
neurons remain viable and responsive to the effects of ketamine anesthesia in patients. Thus, the potential clinical relevance of
in the presence of a space-occupying lesion. In contrast, diffuse the data obtained in rats remains to be clarified. Nevertheless,
intracellular damage occurs in the cytotoxic brain edema, the results of 1 study suggested that the psychotomimetic
CMRO2 may be severely depressed, and autoregulation of the effects of NMDA antagonists in humans may correlate with the
cerebral vasculature may be impaired. As a consequence, the cerebral pathologic changes observed in rats.61 Another crucial
cytotoxic brain becomes less responsive to the vasoactive ef- factor that must be considered when extrapolating results from
fects of intravenous anesthetics, including ketamine. experimental animals to humans is the presence or absence of
The actions of ketamine (10⫺3 to 10⫺7 mol/L) on cerebral ischemia. Clearly, the pathologic conditions during or after
pial arterioles in a closed cranial window model were evaluated cerebral ischemic injury are profoundly different from those
in dogs anesthetized with pentobarbital or isoflurane.40 Neither present in intact, uninjured tissue. Neural ischemia causes
topical nor intravenous (1 mg/kg and 5 mg/kg, respectively) profound increases in extracellular excitatory transmitter con-
ketamine caused pial arteriolar vasodilation. However, the re- centration.62 This observation indirectly suggests that ketamine
activity of pial arterioles to hypercapnia was reduced after may be capable of causing additional neural excitation and
intravenous, but not topical, administration of ketamine, sup- further damage during ischemia-reperfusion injury, but this
porting the hypothesis that the type of baseline anesthetic hypothesis has not been examined.
affects the microvascular effects of ketamine. Ketamine was
also shown to differentially modulate the cerebral circulation in CEREBROVASCULAR AND METABOLIC EFFECTS OF
the presence of volatile agents because ketamine reduced KETAMINE IN HUMAN VOLUNTEERS
isoflurane- but not sevoflurane-induced cerebral vasodilation.41 Incremental intravenous doses of ketamine (cumulative total
of 3 mg/kg) increased CBF, CPP, and arterial CO2 tension and
NEUROTOXIC EFFECTS OF NMDA-RECEPTOR reduced cerebral vascular resistance but did not affect the
ANTAGONISTS: EXPERIMENTAL EVIDENCE CMRO2, CMRglu, and CMRlactate in healthy patients before
In contrast to the aforementioned data, other evidence sug- elective surgery.2 The actions of racemic ketamine (0.25 or 0.5
gests that NMDA-receptor antagonists may produce neurotox- mg/kg intravenously) on spontaneous brain electrical activity
icity under certain experimental conditions. For example, com- and intracranial blood flow velocity were assessed in conscious
plete inhibition of normal NMDA-receptor activity reduced volunteers.63 Ketamine modestly increased heart rate and mean
brain cell survival and worsened physiologic outcome in ro- arterial pressure but did not affect end-tidal CO2 tension or
dents because NMDA-mediated signal transduction was re- arterial O2 saturation. Ketamine also caused a dose-dependent,
quired to express neurotrophins and survival-promoting pro- transient shift in the electroencephalogram (EEG) to synchro-
teins.46 Similarly, large doses of NMDA-receptor antagonists nous, high-voltage slow waves with an increase in total power
caused apoptosis, synaptic deficits, and cognitive impairment in concomitant with increases in middle cerebral artery blood flow
the developing rat brain, especially during the vulnerable phase velocity. These data suggested that enhanced intracranial blood
of synaptogenesis.47-50 Ketamine produced acute, albeit tran- flow velocity occurs primarily as a result of increases in neu-
sient, vacuolar changes in posterior cingulate/retrosplenial cor- ronal activity and not because of changes in hemodynamics,
tices in adult rats.51-53 The coadministration of ketamine and the ventilation, or oxygenation. Subanesthetic doses of ketamine
NMDA antagonist nitrous oxide further enhanced vacuole for- also increased EEG theta activity, regional CBF, and metabo-
mation, whereas the pretreatment with a ␥-aminobutyric acid lism in spontaneously breathing volunteers.63-66 As described
agonist prevented these adverse effects.51,54 Low doses of earlier, these metabolic effects were dependent on the optical
NMDA antagonists, including ketamine and phencyclidine, isomer examined. Vollenweider et al65 investigated the effects
produced reversible pathomorphologic changes in cerebral cor- of (S)- and (R)-ketamine enantiomers on brain energy metab-
tical neurons of the adult rat brain,55 and higher doses of these olism in healthy volunteers using positron emission tomogra-
drugs caused irreversible neuronal degeneration in several cor- phy. Psychotomimetic doses of (S)-ketamine substantially in-
tical-limbic brain regions.54-58 In vitro studies showed cell death creased regional CMRglu in the frontal cortex and thalamus.
in neurons cultured from the rat forebrain after prolonged These metabolic changes were associated with egodisintegra-
exposure to ketamine at concentrations of 10 or 20 ␮mol/L, but tion and hallucinations. In contrast, equimolar doses of (R)-
apoptosis did not occur in the presence of 0.1 or 1.0 ␮mol/L of ketamine suppressed regional CMRglu in the temporomedial
ketamine.59 Studies conducted with cultured monkey frontal cortex and left insula concomitant with a state of relaxation.
cortical neurons produced similar results.60 These results Subanesthetic doses of (S) - and (R)-ketamine were shown to
strongly suggested that neurotoxicity produced by NMDA an- interact differently with the NMDA-and sigma-receptor sites in
tagonists in vitro may be dose-related. the human brain. The sigma receptor was first described as a
The mechanism responsible for NMDA-antagonist neurotox- novel opioid receptor but later was established as a distinct
icity is unclear, but the disinhibition of excitatory neural path- pharmacologic receptor distinguished by its unusual ability to
134
Table 1. Physiologic and Neurocognitive Effects of Ketamine in Humans

Reference Size/Study Group Age Dose, Ketamine Study Setting Outcome

Heart surgery
Tuman et al, 1989 240/345/212/250/47 62/63/63/63/63 3-6 mg/kg Cardiac surgery patients, no NC: neurologic morbidity
FenH/FenM/Sufen/Dia&Ket/Hal cerebral compromise Dia&Ket: D: vasopressors and ECG
evidence of ischemia
Roytblat et al, 1998 14/17 Ket/C 68/65 0.25-mg/kg bolus Cardiac surgery patients, no Ket: D: IL-6 during and post surgery
cerebral compromise
Zilberstein et al, 15/15 Ket/C 64/58 0.25-mg/kg bolus Cardiac surgery patients, no Ket: A: superoxide anion, percentage
2002 cerebral compromise of neutrophils, C: I: superoxide
anion
Nagels et al, 2004 50/56 Ket/Remifen 61/60 2.5-mg/kg bolus and 125 ␮g/kg/ Cardiac surgery patients, no Ket: less D: Trailmaking B test
min cerebral compromise
Smith et al, 2006 21/21 Suf/Ket&Mid 47/47 0.7 mg/kg/min for induction and Cardiac surgery patients, no NC: neurologic morbidity
2 mg/kg/h cerebral compromise Ket&Mid: D: ET-PCO2 Hct and I: MAP
Bartoc et al, 2006 15/18/17 Ket/Ket/C 73/73/69 0.25-, 0.5-mg/kg bolus Cardiac surgery patients, no Ket: D: IL-6 postsurgery in both
cerebral compromise ketamine groups
Ket: D: CRP postsurgery in 0.5 mg/kg
ketamine group
Ket: D: IL-10 postsurgery in both
ketamine groups
Hudetz et al, 2008 24/24 Ket/C 68/67 0.25-mg/kg bolus Cardiac surgery patients, no Ket: less D: delayed figure
cerebral compromise reproduction and delayed word list
recall tests
Hudetz et al, 2009 29/29 Ket/C 68/66 0.25-mg/kg bolus Cardiac surgery patients, no Ket: less delirium
cerebral compromise
Neurology
Mayberg et al, 1995 20 Ket 49 1-mg/kg bolus Neurosurgical patients, NC: MAP, CPP, PaCO2, AVDO2
mildly raised ICP D: VMCA, ICP
Strebel et al, 1995 6/6/6/6 39/43/35/36 2-mg/kg bolus Neurosurgical patients, no C: NC: MAP, VMCA
C/Ket/Ket&Mid/Ket&Esm cerebral compromise Ket: I: VMCA, MAP
Ket&Mid: D:VMCA, NC:MAP
Ket&Esm I: VMCA, NC:MAP
Kolenda et al, 1996 16/17 Ket&Mid/Fen&Mid 38/29 65 mg/kg/d Head-injured patients, Ket&Mid: I: CPP
increased ICP D: vasopressors
Albanese et al, 8 Ket 28 1.5, 3, 5 mg/kg Head-injured patients, NC: CPP, HR VMCA, MAP
1997 increased ICP D: ICP

HUDETZ AND PAGEL


Bourgoin et al, 12/13 Ket&Mid/Suf&Mid 30/27 4.92 ⫾ 1.5 mg Head-injured patients, NC: ICP, CPP
2003 increased ICP Ket&Mid: I: HR
Suf&Mid: I: fluid, vasopressors
NEUROPROTECTION BY KETAMINE
Grathwohl et al, 46/47 Fen/Sufen&Sevo/Iso 27/27 5-20 ␮g/kg/min Head-injured patients, NC: neurologic outcome, death,
2008 Pro&Ket increased ICP injury severity score lowest
intraoperative SBP
General surgery
Roytblat et al, 1996 11/11 Ket/C 51/52 0.15 mg/kg Surgical patients Ket: D: HR, MAP, IL-6
Sakai et al, 2000 7/7/8 Awake/Pro/Pro&Ket 29/33/32 2 mg/kg/h Surgical patients ProKet: NC: VMCA, MAP, HR
Nagase et al, 2001 15/15 Ket/Pro 40/40 1 mg/kg Surgical patients, no Ket: NC: VMCA
cerebral compromise Pro: D: VMCA
Engelhard et al, 12/12 ProKet/Sevo 52/52 2.5 mg/kg/h Surgical patients, no ProKet: NC: Autoregulatory index
2001 cerebral compromise
Beilin et al, 2007 9/10 Ket/C 39/44 0.15 mg/kg Surgical patients, no Ket: D: IL-6, TNF-␣
cerebral compromise
Volunteers
Takeshida et al, 10 Ket 43 2 mg/kg and 1 mg/kg Volunteers, no cerebral I: MAP, CBF, CPP, PaCO2
1972 compromise D: CVR
NC: CMRO2, CMRglucose
CMRlactate
Kochs et al, 1991 10/10 Ket/Ket 27 0.25 mg/kg/0.5 mg/kg Volunteers, no cerebral I: MAP, HR, EEG theta, CBF
compromise NC: ET-PCO2, SaO2
Vollenweider et al, 10 S-Ket/R-Ket 30 15-mg bolus and 0.84-1.2 mg/ Volunteers, no cerebral S-Ket: I: rCMRGlu in anterior
1997 kg/h compromise cingulate, parietal, left
sensorimotor cortex, thalamus
R-Ket: D: rCMRGlu in temporomedial
cortex, left insula
Holcomb et al, 2001 13/10 Ket/C 31/30 0.3-mg/kg bolus Volunteers, no cerebral Ket: I: rCBF in anterior cingulate,
compromise medial frontal, inferior frontal
cortices
Ket: D: rCBF in cerebellum
Langsjo et al, 2003 9 Ket 26 30/100/300 ng/mL Volunteers, no cerebral I: rCBF in anterior cingulate,
compromise thalamus, putamen frontal cortices
NC: rCMRO2

Abbreviations: NC, no change; D, decrease; Ket, ketamine; Remifen, remifentanil; C, control; ICP, intracranial pressure; IL-6, interleukin 6; CRP, C-reactive protein; IL-10, interleukin 10; MAP, mean
arterial pressure; CPP, cerebral perfusion pressure; AVDO2, arteriovenous difference in oxygen content; VMCA, flow velocity in middle cerebral artery; Esm, esmolol; Mid, midazolam; Fen, fentanyl;
HR, heart rate; Pro, propofol; Sevo, sevoflurane; S-Ket, S-ketamine; R-Ket, R-ketamine; I, increase; rCBF, regional cerebral blood flow; rCMRO2, regional cerebral metabolic rate of oxygen; rCBV,
regional cerebral blood volume; rCMRGlu, regional cerebral metabolic rate of glucose; EEG theta, electroencephalogram theta activity; SaO2, arterial oxygen saturation; ET-PCO2, end-tidal
carbondioxid pressure; CVR, cerebral vascular resistance; PaCO2, arterial carbondioxide pressure; CMRO2, cerebral metabolic rate of oxygen; CMRglucose, cerebral metabolic rate of glucose;
CMRlactate, cerebral metabolic rate of lactate; O2, superoxide anion; TNF-␣, tumor necrosis factor ␣; Sufen, sufentanil; Dia, diazepam; FenH, fentanyl high dose; FenM, fentanyl moderate dose; Hal,
halothane; Hct, hematocrit.

135
136 HUDETZ AND PAGEL

bind a wide variety of drugs.67 (S)-ketamine was shown to bind compromised local blood flow. The physiologic effects of
to the phencyclidine binding site of the NMDA receptor with a ketamine in human volunteers are summarized in Table 1.
4- to 5-fold greater affinity than the (R) enantiomer. In contrast,
(R)-ketamine also had a weak affinity, whereas (S)-ketamine CEREBROVASCULAR AND METABOLIC EFFECTS OF
binds only negligibly to the sigma receptor sites.68 This obser- KETAMINE IN SURGICAL PATIENTS
vation allowed the investigators to identify the putative sigma Early studies suggested that ketamine causes parallel in-
effects of (R)-ketamine on CMRglu and mental state. Such a creases in CBF, cerebral blood volume, and CMRO2 and, as a
distinction was important because sigma receptors may modu- result, may produce undesirable increases in ICP in surgical
late glutamatergic neuronal inputs or directly regulate the firing patients.33,75,76 Recent research disputes these long-held suppo-
activity of dopaminergic neurons in schizophrenia.69 Hence, by sitions.77,78 Ventilation was not controlled in many older stud-
modulating the glutamatergic inputs, by directly regulating the ies, and ketamine, especially when combined with propofol or
firing activity of dopaminergic neurons, or by both mecha- midazolam, was shown to reduce ICP and CMRO2 in the
nisms, ketamine with its putative effects on the sigma receptors presence of mechanical ventilation.79 When changes in anterior
could affect the treatment of schizophrenia and therefore play a fontanel pressure (a noninvasive indicator of ICP) were com-
role in neuroprotection. pared during ketamine, fentanyl, isoflurane, or halothane anes-
The temporal pharmacokinetic actions of racemic ketamine thesia in preterm neonates without neurologic disease during
(0.3 mg/kg) on regional CBF were examined using positron mechanical ventilation, ketamine caused similar reductions in
emission tomography in volunteers.66 As observed in experi- anterior fontanel pressure compared with the other drugs.80
mental animals, ketamine produced distinctly different tempo- Kolenda et al81 examined the effects of ketamine on ICP and
ral alterations in regional CBF in specific areas of the human CPP in patients with moderate-to-severe head trauma randomly
brain. Ketamine increased regional CBF in the anterior cingu- assigned to receive ketamine or fentanyl in combination with
late, medial frontal, and inferior frontal cortices but reduced midazolam. In the absence of controlled ventilation, patients
regional CBF in the cerebellum. The peak alteration in regional sedated with ketamine/midazolam required lower quantities of
CBF occurred 6 to 16 minutes after administration of the drug. vasoactive drugs but also had higher ICP and CPP than those
The trend correlations between regional CBF in selected max- receiving fentanyl/midazolam. These results suggested that ket-
ima in frontal cortex/anterior cingulate and behavior suggested amine may be deleterious when used for conscious sedation in
that these simultaneous measurements may be beneficial to link patients with intracranial trauma. In contrast, Mayberg et al78
regional blood flow to function. Thus, ketamine may provide a investigated the effects of ketamine on ICP and CBF velocity in
probe to advance an understanding of psychosis from a dy- isoflurane/nitrous oxide-anesthetized, mechanically-ventilated
namic physiologic perspective70-72 because alterations in re- patients undergoing craniotomy for brain tumor resection or
gional blood flow change patterns may be associated with the cerebral aneurysm clipping. Ketamine (intravenous bolus of 1
appearance of the clinical psychotic symptoms. mg/kg) reduced ICP, middle cerebral artery blood flow veloc-
Langsjo et al64 investigated the neuroprotective effects of ity, and total EEG power, but CPP, O2 extraction, and arterial
racemic ketamine using 15O-labeled water, O2, and carbon CO2 tension were unaffected.78 Racemic ketamine (1.5, 3, or 5
monoxide as positron emission tomography tracers to quantify mg/kg) also significantly decreased ICP in neurosurgical pa-
regional CBF, regional CMRO2, and regional cerebral blood tients with traumatic brain injury during propofol anesthesia
volume, respectively, on selected brain regions in healthy male and mechanical ventilation.79 Again, no differences in CPP,
volunteers. Intravenous infusions of ketamine (targeted to 30, jugular vein bulb O2 saturation, and middle cerebral artery
100, and 300 ng/mL) increased regional CBF in a dose-depen- blood flow velocity were observed between ketamine- and
dent manner. Ketamine increased either absolute or relative placebo-treated patients. Bourgoin et al82 examined and com-
regional CBF in the anterior cingulate, thalamus, putamen, pared the relative safety and efficacy of racemic ketamine and
insula, and frontal cortex. Absolute regional CMRO2 was not midazolam in patients with severe head injury during mechan-
changed; only subtle relative increases in the frontal, parietal, ical ventilation. Neither ketamine/midazolam nor sufentanil/
and occipital cortices and decreases in the cerebellum were midazolam altered ICP or CPP, but patients randomized to
detected. The authors concluded that subanesthetic doses of receive ketamine/midazolam required less aggressive volume
ketamine increase regional CBF but do not alter regional resuscitation early during treatment and also showed a trend
CMRO2, leading to a decrease in regional O2 extraction. De- toward reduced use of vasoactive drugs to maintain arterial
spite the observed dissociation of CBF and CMRO2, uncou- pressure compared with those treated with sufentanil/midazo-
pling of cerebral blood flow and metabolism was unlikely lam. Notably, ketamine/midazolam did not produce adverse
because ketamine previously was shown to increase CMRglu. effects or contribute to more pronounced morbidity or mortal-
Significantly greater increases in regional CBF compared with ity. The authors concluded that ketamine/midazolam sedation
regional CMRO2 have been reported in other studies assessing compares favorably with sufentanil/midazolam in maintaining
the effects of focal neuronal stimulation.73,74 Indeed, alterations ICP and CPP in this vulnerable patient population. Nagase
in regional CBF may more closely follow changes of regional et al83 examined the actions of ketamine and propofol on the
CMRglu than regional CMRO2, suggesting that CBF may be cerebrovascular response to CO2 in mechanically ventilated
regulated for purposes other than O2 delivery for oxidative patients undergoing elective surgery during isoflurane anesthe-
metabolism.74 It may be speculated that the increases in re- sia. Middle cerebral artery blood flow velocity and pulsatility
gional CBF in excess of regional CMRO2 requirements may index assessed using transcranial Doppler were determined
provide a reserve in O2 supply for conditions with possibly under hypo-, normo-, and hypercapnic conditions in the pres-
NEUROPROTECTION BY KETAMINE 137

ence or absence of ketamine (1 mg/kg) or propofol (2 mg/kg ences in the lowest intraoperative systolic blood pressure were
followed by an infusion of 6-10 mg/kg/h). Ketamine, but not observed in patients receiving volatile compared with ket-
propofol, modestly reduced the cerebrovascular response to amine-propofol anesthesia groups. The neurologic outcome
CO2 and prevented increases in middle cerebral artery blood also was similar between groups. These data suggested that
flow velocity during hypercapnia. Such actions may be bene- intravenous propofol-ketamine anesthesia was as efficacious
ficial in patients with increased ICP. Taken together, these data and safe as volatile anesthesia in these severely injured patients.
suggest that ketamine may exert beneficial neurologic effects in The results also confirmed the findings of other studies indi-
anesthetized, mechanically ventilated patients with increased cating that the addition of ketamine to propofol anesthesia
ICP without causing adverse changes in cerebral hemodynam- attenuates the systemic hypotension associated with propofol
ics. The physiologic effects of ketamine in surgical patients are alone by preserving CPP through sympathomimetic effects.88-90
summarized in Table 1. The physiologic effects of ketamine in humans are summarized
in Table 1.
CEREBROVASCULAR AND METABOLIC EFFECTS OF
KETAMINE COMBINED WITH OTHER DRUGS KETAMINE AND NEUROLOGIC IMPAIRMENT AFTER
IN HUMANS CARDIAC SURGERY
The cerebral hemodynamic and metabolic effects of racemic Postoperative neurologic deterioration is common in cardiac
ketamine have been shown to be dependent on the presence of surgical patients, especially when CPB is used. Reduced per-
other anesthetics. Strebel et al84 investigated the actions of fusion pressure during CPB places patients at risk of cerebral
ketamine alone or in combination with midazolam or esmolol ischemia. Embolization of air or particulate matter during aortic
in mechanically ventilated patients anesthetized with isoflu- cannulation or weaning from CPB may produce focal neuro-
rane. Ketamine-induced changes in cerebral hemodynamics logic damage and neuropsychiatric complications.91 CPB also
were measured by using transcranial Doppler ultrasound. As causes a systemic inflammatory response, which may contrib-
expected, ketamine alone increased middle cerebral artery ute to the development of neurologic injury.92,93 This systemic
blood flow velocity and mean arterial pressure compared with inflammation may be mediated by surgical trauma, blood con-
placebo. Ketamine-induced increases in middle cerebral artery tact with the extracorporeal bypass circuit, and lung reperfusion
blood flow velocity were unaffected when mean arterial pres- injury after the discontinuation of CPB. Attenuating this in-
sure was maintained at baseline values by infusion of intrave- flammatory response is an important therapeutic objective and
nous esmolol. In contrast, the deleterious effects of ketamine on is associated with improved outcome.94,95 Pulmonary function
middle cerebral artery blood flow velocity were abolished in may deteriorate significantly after CPB, and this acute lung
the presence of midazolam. The results suggested that midazo- injury also may impair cognitive performance.96 Acute lung
lam profoundly affects the cerebrovascular actions of ketamine injury after CPB was shown to be related to the activation of
in isoflurane-anesthetized patients. The effects of racemic ket- neutrophils and reductions in the body temperature during
amine/propofol anesthesia on intracerebral hemodynamics CPB.97 Neutrophils and the inflammatory mediators that they
were compared with those produced by propofol alone using produce play an important role in the pathogenesis of postop-
transcranial Doppler ultrasound.85 Ketamine/propofol anesthe- erative neurologic dysfunction in cardiac surgery patients.97
sia did not affect heart rate, mean arterial pressure, or middle Lung injury during and after CPB precipitates an accumulation
cerebral artery blood flow velocity. During normocapnic con- of activated neutrophils in the pulmonary circulation with the
ditions, middle cerebral artery blood flow velocity was lower in subsequent release of toxic mediators that contribute to the
propofol-anesthetized patients with or without ketamine com- injury of other vulnerable tissue, including the brain.98,99 Neu-
pared with the conscious state. The cerebral vascular responses trophil elastase may be 1 such mediator of injury.100 Elastase is
to CO2 in patients receiving propofol/ketamine also were sim- a potent protease, capable of damaging intact neurons101 and
ilar to propofol alone. These results suggested that racemic elevated plasma concentrations of elastase that occur in patients
ketamine does not adversely influence middle cerebral artery after CPB.102-105 Notably, the cellular response to the extracor-
blood flow velocity or the cerebrovascular response to CO2 poreal circulation was shown to be delayed after hypothermia,
during propofol anesthesia in patients without intracranial pa- provoking a delayed onset of neutrophilia and elevated plasma
thology. The effects of (S)-ketamine (2.5 mg/kg/h) and propo- pulmonary elastase concentrations.97 Thus, the adverse conse-
fol (target-controlled plasma concentrations of 1.5-2.5 ␮g/mL) quences of CPB may affect postoperative neurologic function.
on cerebrovascular autoregulation were compared with sevo- Ketamine may produce neuroprotective effects in part by
flurane (2.0 minimum alveolar concentration) in patients inhibiting this systemic inflammatory response. Zilberstein et
undergoing elective abdominal surgery.86 Autoregulation was al106 showed that the administration of ketamine (0.25 mg/kg)
maintained during (S)-ketamine/propofol anesthesia but was during anesthetic induction attenuates neutrophil activation in
attenuated during the administration of sevoflurane. Thus, the patients after CPB as indicated by reductions in superoxide
addition of (S)-ketamine to propofol anesthesia does not appear anion production and total postoperative neutrophil count. Ket-
to affect intracranial hemodynamics or autoregulation in surgi- amine also may favorably affect cognitive function and post-
cal patients. surgical inflammation after CPB. Patients undergoing CABG
The effect of anesthetic techniques on neurologic outcomes surgery using CPB were randomized to receive ketamine (0.25
in combat-related traumatic brain injury and the potential ben- mg/kg) or 0.9% saline solution during anesthetic induction with
efits of total intravenous compared with volatile anesthesia fentanyl.107 In contrast to placebo, ketamine produced a sus-
recently were described in a retrospective study.87 No differ- tained suppression of increases in serum IL-6 concentrations
138 HUDETZ AND PAGEL

through the 7th postoperative day. Notably, serum IL-6 con- nitive functions concomitant with reduced C-reactive protein
centrations correlated with the patient’s clinical course after concentrations suggested that the neuroprotective effect of ket-
surgery,107 implying a more beneficial outcome for patients amine may be caused, in part, by an anti-inflammatory action of
receiving ketamine pretreatment compared with those who the drug. Hudetz et al123 also showed that the administration of
did not. The degree of the systemic inflammatory response ketamine (0.5 mg/kg) during anesthetic induction attenuates
has been correlated with patient outcome in cardiac surgical postoperative delirium in a similar patient population. The
patients.108-110 Inflammation also occurs in the brain after incidence of postoperative delirium was significantly lower in
off-pump coronary artery bypass graft surgery111 or after non- patients receiving ketamine (3%) compared with those treated
neurologic, noncardiac surgery, as indicated by increased con- with placebo (31%). Postoperative C-reactive protein concen-
centrations of proinflammatory cytokines in the cerebrospinal tration also was significantly lower in the ketamine-compared
fluid.112 Notably, inflammatory changes in the hippocampus with placebo-treated patients, again suggesting that improve-
may adversely affect learning, memory, and other cognitive ment in short-term neurocognition was related to an anti-
domains.113 The administration of ketamine (0.25 or 0.5 mg/kg) inflammatory effect. Whether these ketamine-induced neuro-
also attenuated increases of serum C-reactive protein, IL-6, and protective effects will translate into long-term benefits remains
IL-10 concentrations during and after cardiac surgery com- to be established, but it is clear that perioperative cognitive
pared with placebo.114 The addition of a single low dose of dysfunction is an important risk factor for long-term disability.
ketamine to the other drugs used for anesthetic induction did The physiologic and neurocognitive effects of ketamine in
not produce adverse hemodynamic effects during induction, cardiac surgical patients are summarized in Table 1.
intubation, incision, or sternotomy, nor did this intervention Preoperative depression is a risk factor for postoperative
contribute to postoperative dysphoria. However, the mean ar- delirium.124,125 Depression may be precipitated by fear of sur-
terial pressure and systemic vascular resistance remained gery and its outcome concomitant with unfamiliar environmen-
higher at the end of surgery, upon arrival in the intensive care tal surroundings.126,127 Limited concentration and attention are
unit, and during the first postoperative day in patients receiving among the most commonly reported symptoms, but impairment
ketamine compared with those who did not receive the disso- in recent verbal and nonverbal memory and impaired executive
ciative anesthetic. In contrast to these intriguing results with functions also have been found in patients with depression.128-131
ketamine, opioids alone did not substantially affect serum C- NMDA-receptor antagonists may be effective for treating de-
reactive protein, IL-6, IL-8, and cortisol concentrations when pression.132 Ketamine (0.5 mg/kg) significantly improved the
used as adjuncts to an inhaled anesthetic.115 Quantitative EEG symptoms of patients with depression within 72 hours after
designed to detect postoperative neurologic injury was similar administration.133 Ketamine (1 mg/kg) also improved postop-
between patients receiving a ketamine-midazolam– based anes- erative depression in patients 1 day after orthopedic surgery.134
thetic compared with those receiving sufentanil anesthesia in Thus, alleviating depression symptoms may reduce cognitive
patients undergoing cardiac surgery with CPB.116 These data impairment after surgery.
suggested that a ketamine-midazolam anesthetic technique Postoperative delirium and postoperative cognitive dysfunc-
does not cause cerebral electrical activation or exacerbate post- tion occur frequently in elderly patients undergoing cardiac
operative neurologic injury. The results of this study116 indi- surgery using CPB. Postoperative delirium often precedes and
rectly supported earlier data by Tuman et al117 showing that may predict the subsequent development of postoperative cog-
ketamine-diazepam anesthesia was associated with decreased nitive dysfunction. To date, few therapeutic options for the
incidences of cerebral ischemia (assessed by EEG) and periop- prevention of postoperative cognitive dysfunction have been
erative hypotension requiring vasoactive drug support com- shown to be effective. Postoperative cognitive dysfunction may
pared with other anesthetic techniques. be observed in as many as three-quarters of cardiac surgical
A neuropsychologic test battery was used to examine and patients at the time of hospital discharge, and these deficits may
compare the potential neuroprotective effects of ketamine and persist in one-third of patients after 6 months.135 Postoperative
remifentanil in patients after cardiac surgery.118 Ketamine im- cognitive dysfunction is characterized by impaired recent mem-
proved cognitive performance in only 1 test (the Trailmaking B ory, concentration, language comprehension, and social inte-
test, a measure of executive functions) compared with remifen- gration.121 Patients with postoperative cognitive dysfunction
tanil. However, interpretation of these results was complicated may experience delayed transfer from the intensive care unit
because propofol, a drug with well-known potent anti-inflam- after surgery, prolonged hospitalization, and a longer recovery
matory properties, was used as a baseline anesthetic, and many before returning to work.136,137 These patients also may expe-
young patients who are not at risk for long-term postoperative rience impaired self-care, increased dependency, increased at-
cognitive dysfunction119-121 were enrolled in the study. In con- trition from rehabilitation, and higher rates of hospital read-
trast to the findings of Nagel et al, the authors recently showed mission. Thus, identifying treatment interventions that
that the administration of ketamine (0.5 mg/kg) during anes- reduce or eliminate postoperative cognitive dysfunction in
thetic induction attenuated short-term postoperative cognitive cardiac surgical patients is an essential goal with direct
dysfunction as indicated by several neuropsychologic tests in clinical implications.
patients anesthetized with isoflurane and fentanyl undergoing
cardiac surgery.122 These findings were accompanied by reduc- CONCLUSION
tions in serum C-reactive protein concentrations on the first and Ketamine has long been considered contraindicated in
third postoperative days in ketamine- compared with placebo- patients with brain injury, but paradoxically this dissociative
treated patients. In this investigation, the preservation of cog- anesthetic may provide beneficial effects in neurologically
NEUROPROTECTION BY KETAMINE 139

impaired patients during controlled ventilation. Ketamine against long-term cognitive dysfunction is currently un-
may exert neuroprotection because the drug inhibits the known. It is also unclear whether ketamine is capable of
NMDA-receptor activation, mediates beneficial changes in exerting neuroprotective effects in other groups of surgical
apoptosis-regulating proteins, and interferes with the inflam- patients who are not subjected to the stress of CPB. Thus, a
matory response to injury when used in typical sedative or clear recommendation for the clinical use of ketamine as a
anesthetic doses. Cardiovascular stimulation by ketamine neuroprotective drug cannot be definitely established at
may also improve cerebral perfusion, and this action may be present. Based on the evidence accumulated to date, an
advantageous in patients after brain injury. Conversely, American Heart Association “class intermediate” recom-
other laboratory investigations have suggested that ketamine mendation for the use of ketamine in cardiac surgery patients
may cause neurotoxicity when administered in larger doses. is warranted. Thus, further research will be required to
Intriguing recent clinical evidence has emerged, suggesting examine the safety, efficacy, and potential limitations of
that ketamine exerts beneficial short-term actions on post- ketamine as a neuroprotective agent before the drug can be
operative delirium and cognitive dysfunction in patients formally recommended for routine clinical use in the cardiac
after cardiac surgery. Whether favorable effects also protect or noncardiac surgical setting.

REFERENCES
1. Messick JM Jr, Casement B, Sharbrough FW, et al: Correlation of 17. Barone FC, Arvin B, White RF, et al: Tumor necrosis factor-
regional cerebral blood flow (rCBF) with EEG changes during isoflu- alpha. A mediator of focal ischemic brain injury. Stroke 28:1233-1244,
rane anesthesia for carotid endarterectomy: critical rCBF. Anesthesi- 1997
ology 66:344-349, 1987 18. Himmelseher S, Pfenninger E, Kochs E, et al: S(⫹)-ketamine
2. Takeshita H, Okuda Y, Sari A: The effects of ketamine on up-regulates neuronal regeneration associated proteins following glu-
cerebral circulation and metabolism in man. Anesthesiology 36:69-75, tamate injury in cultured rat hippocampal neurons. J Neurosurg Anes-
1972 thesiol 12:84-94, 2000
3. Dirnagl U, Iadecola C, Moskowitz MA: Pathobiology of isch- 19. Gonzales JM, Loeb AL, Reichard PS, et al: Ketamine inhibits
aemic stroke: An integrated view. Trends Neurosci 22:391-397, 1999 glutamate-, N-methyl-D-aspartate-, and quisqualate-stimulated cGMP
production in cultured cerebral neurons. Anesthesiology 82:205-213,
4. Martin RL, Lloyd HG, Cowan AI: The early events of oxygen and
1995
glucose deprivation: setting the scene for neuronal death? Trends
20. Orser BA, Pennefather PS, MacDonald JF: Multiple mecha-
Neurosci 17:251-257, 1994
nisms of ketamine blockade of N-methyl-D-aspartate receptors. Anes-
5. Katsura K, Kristian T, Siesjo BK: Energy metabolism, ion ho-
thesiology 86:903-917, 1997
meostasis, and cell damage in the brain. Biochem Soc Trans 22:991-
21. Garthwaite J: Glutamate, nitric oxide and cell-cell signalling in
996, 1994
the nervous system. Trends Neurosci 14:60-67, 1991
6. Meldrum BS: Glutamate as a neurotransmitter in the brain: Re- 22. Proescholdt M, Heimann A, Kempski O: Neuroprotection of
view of physiology and pathology. J Nutr 130:1007S-1015S, 2000 S(⫹) ketamine isomer in global forebrain ischemia. Brain Res 904:
7. Leist M, Nicotera P: Apoptosis, excitotoxicity, and neuropathol- 245-251, 2001
ogy. Exp Cell Res 239:183-201, 1998 23. Engelhard K, Werner C, Eberspacher E, et al: The effect of the
8. Prass K, Dirnagl U: Glutamate antagonists in therapy of stroke. alpha 2-agonist dexmedetomidine and the N-methyl-D-aspartate antag-
Restor Neurol Neurosci 13:3-10, 1998 onist S(⫹)-ketamine on the expression of apoptosis-regulating proteins
9. Himmelseher S, Pfenninger E, Georgieff M: The effects of ket- after incomplete cerebral ischemia and reperfusion in rats. Anesth
amine-isomers on neuronal injury and regeneration in rat hippocampal Analg 96:524-531, 2003
neurons. Anesth Analg 83:505-512, 1996 24. Kawasaki T, Ogata M, Kawasaki C, et al: Ketamine suppresses
10. O’Neill LA, Kaltschmidt C: NF-kappa B: A crucial transcription proinflammatory cytokine production in human whole blood in vitro.
factor for glial and neuronal cell function. Trends Neurosci 20:252-258, Anesth Analg 89:665-669, 1999
1997 25. Szekely A, Heindl B, Zahler S, et al: S(⫹)-ketamine, but not
11. Shen C, Nettleton D, Jiang M, et al: Roles of the HIF-1 hypoxia- R(⫺)-ketamine, reduces postischemic adherence of neutrophils in the
inducible factor during hypoxia response in Caenorhabditis elegans. coronary system of isolated guinea pig hearts. Anesth Analg 88:1017-
J Biol Chem 280:20580-20588, 2005 1024, 1999
26. Shapira Y, Artru AA, Lam AM: Ketamine decreases cerebral
12. Sakai T, Ichiyama T, Whitten CW, et al: Ketamine suppresses
infarct volume and improves neurological outcome following experi-
endotoxin-induced NF-kappaB expression. Can J Anaesth 47:1019-
mental head trauma in rats. J Neurosurg Anesthesiol 4:231-240, 1992
1024, 2000
27. Miyamoto E, Nakao S, Tomimoto H, et al: Ketamine attenuates
13. Baldwin AS Jr: The NF-kappa B and I kappa B proteins: New
hypocapnia-induced neuronal damage in the caudoputamen in a rat
discoveries and insights. Annu Rev Immunol 14:649-683, 1996
model of chronic cerebral hypoperfusion. Neurosci Lett 354:26-29,
14. del Zoppo GJ, Schmid-Schonbein GW, Mori E, et al: Polymor- 2004
phonuclear leukocytes occlude capillaries following middle cerebral 28. Hoffman WE, Pelligrino D, Werner C, et al: Ketamine decreases
artery occlusion and reperfusion in baboons. Stroke 22:1276-1283, plasma catecholamines and improves outcome from incomplete cere-
1991 bral ischemia in rats. Anesthesiology 76:755-762, 1992
15. Hudetz AG, Oliver JA, Wood JD, et al: Leukocyte adhesion in 29. Church J, Zeman S, Lodge D: The neuroprotective action of
pial cerebral venules after PMA stimulation and ischemia/reperfusion ketamine and MK-801 after transient cerebral ischemia in rats. Anes-
in vivo. Adv Exp Med Biol 411:513-518, 1997 thesiology 69:702-709, 1988
16. Hudetz AG, Wood JD, Kampine JP: Nitric oxide synthase 30. Dawson B, Michenfelder JD, Theye RA: Effects of ketamine on
inhibitor augments post-ischemic leukocyte adhesion in the cerebral canine cerebral blood flow and metabolism: Modification by prior
microcirculation in vivo. Neurol Res 21:378-384, 1999 administration of thiopental. Anesth Analg 50:443-447, 1971
140 HUDETZ AND PAGEL

31. Oren RE, Rasool NA, Rubinstein EH: Effect of ketamine on 51. Jevtovic-Todorovic V, Benshoff N, Olney JW: Ketamine poten-
cerebral cortical blood flow and metabolism in rabbits. Stroke 18:441- tiates cerebrocortical damage induced by the common anaesthetic agent
444, 1987 nitrous oxide in adult rats. Br J Pharmacol 130:1692-1698, 2000
32. Cavazzuti M, Porro CA, Biral GP, et al: Ketamine effects on 52. Jevtovic-Todorovic V, Wozniak DF, Benshoff ND, et al: A
local cerebral blood flow and metabolism in the rat. J Cereb Blood comparative evaluation of the neurotoxic properties of ketamine and
Flow Metab 7:806-811, 1987 nitrous oxide. Brain Res 895:264-267, 2001
33. Sakabe TN: Effects of Anesthetic Agents and Other Drugs on 53. Nakao S, Miyamoto E, Masuzawa M, et al: Ketamine-induced
Cerebral Blood Flow, Metabolism, and Intracranial Pressure. St Louis, c-Fos expression in the mouse posterior cingulate and retrosplenial
MO, Mosby, 2001 cortices is mediated not only via NMDA receptors but also via sigma
34. Schwedler M, Miletich DJ, Albrecht RF: Cerebral blood flow receptors. Brain Res 926:191-196, 2002
and metabolism following ketamine administration. Can Anaesth Soc J 54. Olney JW, Labruyere J, Wang G, et al: NMDA antagonist
29:222-226, 1982 neurotoxicity: Mechanism and prevention. Science 254:1515-1518,
35. Akeson J, Bjorkman S, Messeter K, et al: Cerebral pharmaco- 1991
dynamics of anaesthetic and subanaesthetic doses of ketamine in the 55. Olney JW, Labruyere J, Price MT: Pathological changes induced
normoventilated pig. Acta Anaesthesiol Scand 37:211-218, 1993 in cerebrocortical neurons by phencyclidine and related drugs. Science
36. Corssen G, Miyasaka M, Domino EF: Changing concepts in 244:1360-1362, 1989
pain control during surgery: Dissociative anesthesia with CI-581. A 56. Fix AS, Horn JW, Wightman KA, et al: Neuronal vacuolization
progress report. Anesth Analg 47:746-759, 1968 and necrosis induced by the noncompetitive N-methyl-D-aspartate
37. Mori K, Kawamata M, Mitani H, et al: A neurophysiologic (NMDA) antagonist MK(⫹)801 (dizocilpine maleate): A light and
study of ketamine anesthesia in the cat. Anesthesiology 35:373-383, electron microscopic evaluation of the rat retrosplenial cortex. Exp
1971 Neurol 123:204-215, 1993
38. Kayama Y, Iwama K: The EEG, evoked potentials, and single- 57. Corso TD, Sesma MA, Tenkova TI, et al: Multifocal brain
unit activity during ketamine anesthesia in cats. Anesthesiology 36: damage induced by phencyclidine is augmented by pilocarpine. Brain
316-328, 1972 Res 752:1-14, 1997
39. Nimkoff L, Quinn C, Silver P, et al: The effects of intravenous 58. Farber NB, Wozniak DF, Price MT, et al: Age-specific neuro-
anesthetics on intracranial pressure and cerebral perfusion pressure in toxicity in the rat associated with NMDA receptor blockade: Potential
two feline models of brain edema. J Crit Care 12:132-136, 1997 relevance to schizophrenia? Biol Psychiatry 38:788-796, 1995
40. Ohata H, Iida H, Nagase K, et al: The effects of topical and 59. Wang C, Sadovova N, Fu X, et al: The role of the N-methyl-
intravenous ketamine on cerebral arterioles in dogs receiving pento- D-aspartate receptor in ketamine-induced apoptosis in rat forebrain
barbital or isoflurane anesthesia. Anesth Analg 93:697-702, 2001 culture. Neuroscience 132:967-977, 2005
41. Nagase K, Iida H, Dohi S: Effects of ketamine on isoflurane- and 60. Wang C, Sadovova N, Hotchkiss C, et al: Blockade of N-
sevoflurane-induced cerebral vasodilation in rabbits. J Neurosurg An- methyl-D-aspartate receptors by ketamine produces loss of postnatal
esthesiol 15:98-103, 2003 day 3 monkey frontal cortical neurons in culture. Toxicol Sci 91:192-
42. Wendling WW, Chen D, Daniels FB, et al: The effects of 201, 2006
N-methyl-D-aspartate agonists and antagonists on isolated bovine ce- 61. Anand KJ, Soriano SG: Anesthetic agents and the immature
rebral arteries. Anesth Analg 82:264-268, 1996 brain: Are these toxic or therapeutic? Anesthesiology 101:527-530,
43. Shakunaga K, Kojima S, Jomura K, et al: Ketamine suppresses 2004
the production and release of endothelin 1 from cultured bovine endo- 62. Arundine M, Tymianski M: Molecular mechanisms of gluta-
thelial cells. Anesth Analg 86:1098-1102, 1998 mate-dependent neurodegeneration in ischemia and traumatic brain
44. Nagase K, Iida H, Dohi S: L-arginine and nitroglycerin restore injury. Cell Mol Life Sci 61:657-668, 2004
hypercapnia-induced cerebral vasodilation in rabbits after its attenua- 63. Kochs E, Werner C, Hoffman WE, et al: Concurrent increases in
tion by ketamine. Anesth Analg 94:954-958, 2002 brain electrical activity and intracranial blood flow velocity during
45. Chi OZ, Wei HM, Klein SL, et al: Effect of ketamine on low-dose ketamine anaesthesia. Can J Anaesth 38:826-830, 1991
heterogeneity of cerebral microregional venous O2 saturation in the rat. 64. Langsjo JW, Kaisti KK, Aalto S, et al: Effects of subanesthetic
Anesth Analg 79:860-866, 1994 doses of ketamine on regional cerebral blood flow, oxygen consump-
46. Hardingham GE, Fukunaga Y, Bading H: Extrasynaptic tion, and blood volume in humans. Anesthesiology 99:614-623, 2003
NMDARs oppose synaptic NMDARs by triggering CREB shut-off and 65. Vollenweider FX, Leenders KL, Oye I, et al: Differential psy-
cell death pathways. Nat Neurosci 5:405-414, 2002 chopathology and patterns of cerebral glucose utilisation produced by
47. Ikonomidou C, Bosch F, Miksa M, et al: Blockade of NMDA (S)- and (R)-ketamine in healthy volunteers using positron emission
receptors and apoptotic neurodegeneration in the developing brain. tomography (PET). Eur Neuropsychopharmacol 7:25-38, 1997
Science 283:70-74, 1999 66. Holcomb HH, Lahti AC, Medoff DR, et al: Sequential regional
48. Hayashi H, Dikkes P, Soriano SG: Repeated administration of cerebral blood flow brain scans using PET with H2(15)O demonstrate
ketamine may lead to neuronal degeneration in the developing rat brain. ketamine actions in CNS dynamically. Neuropsychopharmacology 25:
Paediatr Anaesth 12:770-774, 2002 165-172, 2001
49. Jevtovic-Todorovic V, Hartman RE, Izumi Y, et al: Early ex- 67. Martin WR, Eades CG, Thompson JA, et al: The effects of
posure to common anesthetic agents causes widespread neurodegen- morphine- and nalorphine- like drugs in the nondependent and mor-
eration in the developing rat brain and persistent learning deficits. phine-dependent chronic spinal dog. J Pharmacol Exp Ther 197:517-
J Neurosci 23:876-882, 2003 532, 1976
50. Scallet AC, Schmued LC, Slikker W Jr, et al: Developmental 68. Oye I, Hustveit O, Maurset A, et al: The ciral forms of ketamine
neurotoxicity of ketamine: Morphometric confirmation, exposure pa- as probes for NMDA-receptor function in humans, in Kameyama T,
rameters, and multiple fluorescent labeling of apoptotic neurons. Toxi- Nabeshima T, Domino EF (eds): NMDA Receptor Related Agents.
col Sci 81:364-370, 2004 Ann Arbor, MI, NPP Books, 1991
NEUROPROTECTION BY KETAMINE 141

69. Debonnel G: Current hypotheses on sigma receptors and their 89. Furuya A, Matsukawa T, Ozaki M, et al: Intravenous ketamine
physiological role: possible implications in psychiatry. J Psychiatry attenuates arterial pressure changes during the induction of anaesthesia
Neurosci 18:157-172, 1993 with propofol. Eur J Anaesthesiol 18:88-92, 2001
70. Krystal JH, Karper LP, Seibyl JP, et al: Subanesthetic effects of 90. Wong DH, Jenkins LC: The cardiovascular effects of ketamine
the noncompetitive NMDA antagonist, ketamine, in humans. Psychot- in hypotensive states. Can Anaesth Soc J 22:339-348, 1975
omimetic, perceptual, cognitive, and neuroendocrine responses. Arch 91. Slogoff S, Girgis KZ, Keats AS: Etiologic factors in neuropsy-
Gen Psychiatry 51:199-214, 1994 chiatric complications associated with cardiopulmonary bypass. Anesth
71. Lahti AC, Koffel B, LaPorte D, et al: Subanesthetic doses of Analg 61:903-911, 1982
ketamine stimulate psychosis in schizophrenia. Neuropsychopharma- 92. Laffey JG, Boylan JF, Cheng DC: The systemic inflammatory
cology 13:9-19, 1995 response to cardiac surgery: Implications for the anesthesiologist. An-
72. Malhotra AK, Pinals DA, Weingartner H, et al: NMDA receptor esthesiology 97:215-252, 2002
function and human cognition: The effects of ketamine in healthy 93. Butler J, Rocker GM, Westaby S: Inflammatory response to
volunteers. Neuropsychopharmacology 14:301-307, 1996 cardiopulmonary bypass. Ann Thorac Surg 55:552-559, 1993
94. Zanetti G, Heumann D, Gerain J, et al: Cytokine production
73. Fox PT, Raichle ME: Focal physiological uncoupling of cerebral
after intravenous or peritoneal gram-negative bacterial challenge in
blood flow and oxidative metabolism during somatosensory stimulation
mice. Comparative protective efficacy of antibodies to tumor necrosis
in human subjects. Proc Natl Acad Sci U S A 83:1140-1144, 1986
factor-alpha and to lipopolysaccharide. J Immunol 148:1890-1897,
74. Fox PT, Raichle ME, Mintun MA, et al: Nonoxidative glucose
1992
consumption during focal physiologic neural activity. Science 241:462-
95. Giroir BP: Mediators of septic shock: New approaches for
464, 1988
interrupting the endogenous inflammatory cascade. Crit Care Med
75. Madson JBC: The Effects of Anesthetics Upon Cerebral Circu- 21:780-789, 1993
lation and Metabolism: Experimental and Clinical Studies. New York, 96. Liesker JJ, Postma DS, Beukema RJ, et al: Cognitive perfor-
NY, Springer, 1990 mance in patients with COPD. Respir Med 98:351-356, 2004
76. Wyte SR, Shapiro HM, Turner P, et al: Ketamine-induced 97. Tonz M, Mihaljevic T, von Segesser LK, et al: Acute lung injury
intracranial hypertension. Anesthesiology 36:174-176, 1972 during cardiopulmonary bypass. Are the neutrophils responsible? Chest
77. Himmelseher S, Durieux ME: Revising a dogma: Ketamine for 108:1551-1556, 1995
patients with neurological injury? Anesth Analg 101:524-534, 2005 98. Fountain SW, Martin BA, Musclow CE, et al: Pulmonary leu-
78. Mayberg TS, Lam AM, Matta BF, et al: Ketamine does not kostasis and its relationship to pulmonary dysfunction in sheep and
increase cerebral blood flow velocity or intracranial pressure during rabbits. Circ Res 46:175-180, 1980
isoflurane/nitrous oxide anesthesia in patients undergoing craniotomy. 99. Bando K, Pillai R, Cameron DE, et al: Leukocyte depletion
Anesth Analg 81:84-89, 1995 ameliorates free radical-mediated lung injury after cardiopulmonary
79. Albanese J, Arnaud S, Rey M, et al: Ketamine decreases intra- bypass. J Thorac Cardiovasc Surg 99:873-877, 1990
cranial pressure and electroencephalographic activity in traumatic brain 100. Havemann K, Gramse M: Physiology and pathophysiology of
injury patients during propofol sedation. Anesthesiology 87:1328- neutral proteinases of human granulocytes. Adv Exp Med Biol 167:1-
1334, 1997 20, 1984
80. Friesen RH, Thieme RE, Honda AT, et al: Changes in anterior 101. Janoff A: Elastase in tissue injury. Annu Rev Med 36:207-216,
fontanel pressure in preterm neonates receiving isoflurane, halothane, 1985
fentanyl, or ketamine. Anesth Analg 66:431-434, 1987 102. Riegel W, Spillner G, Schlosser V, et al: Plasma levels of main
81. Kolenda H, Gremmelt A, Rading S, et al: Ketamine for analgo- granulocyte components during cardiopulmonary bypass. J Thorac
sedative therapy in intensive care treatment of head-injured patients. Cardiovasc Surg 95:1014-1019, 1988
Acta Neurochir (Wien) 138:1193-1199, 1996 103. Butler J, Parker D, Pillai R, et al: Effect of cardiopulmonary
82. Bourgoin A, Albanese J, Wereszczynski N, et al: Safety of bypass on systemic release of neutrophil elastase and tumor necrosis
sedation with ketamine in severe head injury patients: Comparison with factor. J Thorac Cardiovasc Surg 105:25-30, 1993
sufentanil. Crit Care Med 31:711-717, 2003 104. Finn A, Naik S, Klein N, et al: Interleukin-8 release and
neutrophil degranulation after pediatric cardiopulmonary bypass.
83. Nagase K, Iida H, Ohata H, et al: Ketamine, not propofol,
J Thorac Cardiovasc Surg 105:234-241, 1993
attenuates cerebrovascular response to carbon dioxide in humans with
105. Faymonville ME, Pincemail J, Duchateau J, et al: Myeloper-
isoflurane anesthesia. J Clin Anesth 13:551-555, 2001
oxidase and elastase as markers of leukocyte activation during cardio-
84. Strebel S, Kaufmann M, Maitre L, et al: Effects of ketamine on
pulmonary bypass in humans. J Thorac Cardiovasc Surg 102:309-317,
cerebral blood flow velocity in humans. Influence of pretreatment with 1991
midazolam or esmolol. Anaesthesia 50:223-228, 1995
106. Zilberstein G, Levy R, Rachinsky M, et al: Ketamine attenuates
85. Sakai K, Cho S, Fukusaki M, et al: The effects of propofol with neutrophil activation after cardiopulmonary bypass. Anesth Analg 95:
and without ketamine on human cerebral blood flow velocity and 531-536, 2002
CO(2) response. Anesth Analg 90:377-382, 2000 107. Roytblat L, Talmor D, Rachinsky M, et al: Ketamine attenuates
86. Engelhard K, Werner C, Mollenberg O, et al: S(⫹)-ketamine/ the interleukin-6 response after cardiopulmonary bypass. Anesth Analg
propofol maintain dynamic cerebrovascular autoregulation in humans. 87:266-271, 1998
Can J Anaesth 48:1034-1039, 2001 108. Cremer J, Martin M, Redl H, et al: Systemic inflammatory
87. Grathwohl KW, Black IH, Spinella PC, et al: Total intravenous response syndrome after cardiac operations. Ann Thorac Surg 61:1714-
anesthesia including ketamine versus volatile gas anesthesia for com- 1720, 1996
bat-related operative traumatic brain injury. Anesthesiology 109:44-53, 109. Deng MC, Dasch B, Erren M, et al: Impact of left ventricular
2008 dysfunction on cytokines, hemodynamics, and outcome in bypass graft-
88. Frizelle HP, Duranteau J, Samii K: A comparison of propofol ing. Ann Thorac Surg 62:184-190, 1996
with a propofol-ketamine combination for sedation during spinal an- 110. Hennein HA, Ebba H, Rodriguez JL, et al: Relationship of the
esthesia. Anesth Analg 84:1318-1322, 1997 proinflammatory cytokines to myocardial ischemia and dysfunction
142 HUDETZ AND PAGEL

after uncomplicated coronary revascularization. J Thorac Cardiovasc 124. Greene N, Attix D, Macy S, et al: Preoperative Executive
Surg 108:626-635, 1994 Function and Depression Predict Postoperaive Delirium. Orlando, FL,
111. Kalman J, Juhasz A, Bogats G, et al: Elevated levels of in- American Society of Anesthesiologists, 2008, p A842
flammatory biomarkers in the cerebrospinal fluid after coronary artery 125. Rudolph JL, Jones RN, Levkoff SE, et al: Derivation and
bypass surgery are predictors of cognitive decline. Neurochem Int validation of a preoperative prediction rule for delirium after cardiac
48:177-180, 2006 surgery. Circulation 119:229-236, 2008
112. Buvanendran A, Kroin JS, Berger RA, et al: Upregulation of 126. Stump DA: Selection and clinical significance of neuropsycho-
prostaglandin E2 and interleukins in the central nervous system and logic tests. Ann Thorac Surg 59:1340-1344, 1995
peripheral tissue during and after surgery in humans. Anesthesiology 127. Jones MJ, Piggott SE, Vaughan RS, et al: Cognitive and
104:403-410, 2006 functional competence after anaesthesia in patients aged over 60:
113. Dantzer R, Kelley KW: Twenty years of research on cytokine- Controlled trial of general and regional anaesthesia for elective hip or
induced sickness behavior. Brain Behav Immun 21:153-160, 2007 knee replacement. BMJ 300:1683-1687, 1990
114. Bartoc C, Frumento RJ, Jalbout M, et al: A randomized, 128. Landro NI, Stiles TC, Sletvold H: Neuropsychological function
double-blind, placebo-controlled study assessing the anti-inflammatory in nonpsychotic unipolar major depression. Neuropsychiatry Neuro-
effects of ketamine in cardiac surgical patients. J Cardiothorac Vasc psychol Behav Neurol 14:233-240, 2001
Anesth 20:217-222, 2006 129. Rose EJ, Ebmeier KP: Pattern of impaired working memory
115. Taylor NM, Lacoumenta S, Hall GM: Fentanyl and the inter- during major depression. J Affect Disord 90:149-161, 2006
leukin-6 response to surgery. Anaesthesia 52:112-115, 1997 130. MacQueen GM, Galway TM, Hay J, et al: Recollection mem-
116. Smith FJ, Bartel PR, Hugo JM, et al: Anesthetic technique ory deficits in patients with major depressive disorder predicted by past
(sufentanil versus ketamine plus midazolam) and quantitative electro- depressions but not current mood state or treatment status. Psychol Med
encephalographic changes after cardiac surgery. J Cardiothorac Vasc 32:251-258, 2002
Anesth 20:520-525, 2006 131. Taylor Tavares JV, Clark L, Cannon DM, et al: Distinct pro-
117. Tuman KJ, McCarthy RJ, Spiess BD, et al: Does choice of files of neurocognitive function in unmedicated unipolar depression
anesthetic agent significantly affect outcome after coronary artery and bipolar II depression. Biol Psychiatry 62:917-924, 2007
surgery? Anesthesiology 70:189-198, 1989 132. Skolnick P, Layer RT, Popik P, et al: Adaptation of N-methyl-
118. Nagels W, Demeyere R, Van Hemelrijck J, et al: Evaluation of D-aspartate (NMDA) receptors following antidepressant treatment: Im-
the neuroprotective effects of S(⫹)-ketamine during open-heart sur- plications for the pharmacotherapy of depression. Pharmacopsychiatry
gery. Anesth Analg 98:1595-1603, 2004 29:23-26, 1996
119. Marcantonio ER, Juarez G, Goldman L, et al: The relationship 133. Berman RM, Cappiello A, Anand A, et al: Antidepressant
of postoperative delirium with psychoactive medications. JAMA 272: effects of ketamine in depressed patients. Biol Psychiatry 47:351-354,
1518-1522, 1994 2000
120. Dyer CB, Ashton CM, Teasdale TA: Postoperative delirium. A 134. Kudoh A, Takahira Y, Katagai H, et al: Small-dose ketamine
review of 80 primary data-collection studies. Arch Intern Med 155: improves the postoperative state of depressed patients. Anesth Analg
461-465, 1995 95:114-118, 2002
121. Moller JT, Cluitmans P, Rasmussen LS, et al: Long-term 135. Newman MF, Kirchner JL, Phillips-Bute B, et al: Longitudinal
postoperative cognitive dysfunction in the elderly ISPOCD1 study. assessment of neurocognitive function after coronary-artery bypass
ISPOCD investigators. International Study of Post-Operative Cognitive surgery. N Engl J Med 344:395-402, 2001
Dysfunction. Lancet 351:857-861, 1998 136. Wilmore DW, Kehlet H: Management of patients in fast track
122. Hudetz JA, Iqbal Z, Gandhi SD, et al: Ketamine attenuates surgery. BMJ 322:473-476, 2001
postoperative cognitive dysfunction one week after cardiac surgery. 137. Roach GW, Kanchuger M, Mangano CM, et al: Adverse ce-
Anesthesiology 109:A826, 2008 rebral outcomes after coronary bypass surgery. Multicenter Study of
123. Hudetz JA, Patterson KM, Iqbal Z, et al: Ketamine attenuates Perioperative Ischemia Research Group and the Ischemia Research and
delirium after cardiac surgery with cardiopulmonary bypass. J Cardio- Education Foundation Investigators. N Engl J Med 335:1857-1863,
thorac Vasc Anesth 2009 Feb 19 [Epub ahead of print] 1996

Anda mungkin juga menyukai