Anda di halaman 1dari 17

Articles

Mechanisms of action, physiological effects, and complications of


hypothermia
Kees H. Polderman, MD

Background: Mild to moderate hypothermia (32–35°C) is the tance, impaired drug clearance, and mild coagulopathy. Targeted
first treatment with proven efficacy for postischemic neurological interventions are required to effectively manage these side ef-
injury. In recent years important insights have been gained into fects. Hypothermia does not decrease myocardial contractility or
the mechanisms underlying hypothermia’s protective effects; in induce hypotension if hypovolemia is corrected, and preliminary
addition, physiological and pathophysiological changes associ- evidence suggests that it can be safely used in patients with
ated with cooling have become better understood. cardiac shock. Cardiac output will decrease due to hypothermia-
Objective: To discuss hypothermia’s mechanisms of action, to induced bradycardia, but given that metabolic rate also decreases
review (patho)physiological changes associated with cooling, and the balance between supply and demand, is usually maintained or
to discuss potential side effects. improved. In contrast to deep hypothermia (<30°C), moderate
Design: Review article. hypothermia does not induce arrhythmias; indeed, the evidence
Interventions: None. suggests that arrhythmias can be prevented and/or more easily
Main Results: A myriad of destructive processes unfold in treated under hypothermic conditions.
injured tissue following ischemia–reperfusion. These include ex- Conclusions: Therapeutic hypothermia is a highly promising
citotoxicty, neuroinflammation, apoptosis, free radical production, treatment, but the potential side effects need to be properly
seizure activity, blood– brain barrier disruption, blood vessel leak- managed particularly if prolonged treatment periods are required.
age, cerebral thermopooling, and numerous others. The severity Understanding the underlying mechanisms, awareness of physi-
of this destructive cascade determines whether injured cells will ological changes associated with cooling, and prevention of po-
survive or die. Hypothermia can inhibit or mitigate all of these tential side effects are all key factors for its effective clinical
mechanisms, while stimulating protective systems such as early usage. (Crit Care Med 2009; 37[Suppl.]:S186 –S202)
gene activation. Hypothermia is also effective in mitigating intra- KEY WORDS: hypothermia; normothermia; fever; mechanisms;
cranial hypertension and reducing brain edema. Side effects neuroprotection; side effects; neurological injury; cardiac arrest;
include immunosuppression with increased infection risk, cold traumatic brain injury
diuresis and hypovolemia, electrolyte disorders, insulin resis-

W hen applying hypother- erate-deep (30°C–31.9°C), and deep patient, because the available cooling and
mia in a clinical setting, it (⬍30°C) hypothermia (1). Insufficient rewarming methods were not very reli-
is important to be aware understanding of these mechanisms is able. The most frequently used cooling
of the underlying mecha- likely to decrease therapeutic efficacy, methods were placement of slabs of ice,
nisms through which the treatment ex- and can at worst lead to treatment fail- ice pads, and refrigerated water on the
erts its effects. Understanding these is- ure. This is illustrated by early experi- patient’s skin, and sometimes the open-
sues can help guide clinical decisions ences with induced hypothermia in the ing of windows of the hospital wards dur-
regarding the required depth and dura- treatment of cardiac arrest and traumatic ing the winter months (to the disconcert-
tion of the treatment, and help in under- brain injury (TBI), and other indications ment of some members of the medical
standing physiological changes and side in the 1940s, 1950s, and 1960s (2–15). At and nursing staff) (11). Because intensive
effects occurring during mild (34°C– that time, it was presumed that hypother- care facilities did not become widely
35.9°C), moderate (32°C–33.9°C), mod- mia exerted its effects exclusively available until the early 1960s, the treat-
through temperature-dependent reduc- ment initially had to be applied in general
tions in metabolism, leading to decreased wards (2–11). Duration of cooling varied
From the Clinical Research, Investigation, and Sys- demand for oxygen and glucose in the considerably, ranging from 2 to 3 days to
tems Modeling of Acute Illness (CRISMA) Laboratory, brain. Patients were routinely treated up to 10 days.
Department of Critical Care Medicine, University of
Pittsburgh School of Medicine, Pittsburgh, United with deep hypothermia (⬍30°C), based Despite this lack of well-controllable
States. on the (erroneous) assumption that this cooling methods and the use of temper-
The author has not disclosed any potential con- was the sole mechanism of action, and atures ⬍30°C (with a much greater risk
flicts of interest. that large reductions in temperature for severe side effects compared with
For information regarding this article, E-mail:
k.polderman@tip.nl or poldermankh@ccm.upmc.edu were required to achieve efficacy. In ad- temperatures ⱖ30°C), many of these
Copyright © 2009 by the Society of Critical Care dition, core temperatures that were actu- studies reported benefits compared with
Medicine and Lippincott Williams & Wilkins ally achieved varied considerably both be- “expected outcome” or historical controls
DOI: 10.1097/CCM.0b013e3181aa5241 tween patients and within the same (2–11). However, these benefits were

S186 Crit Care Med 2009 Vol. 37, No. 7 (Suppl.)


variable and uncertain, and as is to be The side effects of hypothermia can be far role in injury development in a particular
expected there were significant problems more easily managed in this setting, and animal model, whereas this particular
in patient management. In the late 1960s, the methods for inducing and maintain- mechanism is far less important in hu-
interest among clinicians in therapeutic ing hypothermia have improved signifi- mans (25–27). Such a treatment would
usage of hypothermia seems to have sim- cantly since the 1950s. These new in- be highly effective in that animal model,
ply fizzled out; there is a 38-yr gap be- sights and improvements in facilities but would prove far less effective in a
tween the publication of the first two case have set the stage for a successful come- clinical trial.
series describing use of hypothermia in back of hypothermia as a clinical treat- Such differences may result in overes-
cardiac arrest patients and the next study ment. timation of the potential positive effects
(8, 9, 16). These experiences illustrate the Studies on mechanisms underlying of hypothermia; the brains of some ani-
difficulties that can be encountered dur- hypothermia’s protective effects point to mals, especially small animals such as
ing use of controlled hypothermia. four key factors determining success or rodents, appear to be more responsive to
Interest in cooling was rekindled in failure of cooling treatment. These are: neuroprotection than the human brain
the early 1980s by the positive results (28, 29). On the other hand, protective
from numerous animal experiments, ● Speed of induction of hypothermia; effects may also be underestimated when
some of which are discussed below. Many outcomes in animal experiments are looking at only one mechanism and/or
important insights regarding effective far better when cooling is initiated rap- only one animal model. For example,
use of hypothermia and the mechanisms idly after injury. many animal studies have focused on the
underlying its protective effects were ● Duration of cooling (depending on the so-called neuroexcitotoxic cascade,
gained at that time. An important break- severity of the initial injury and the which is central to the development of
through was the realization that neuro- time interval until target temperature postischemic injury in many primitive
logical outcome could be improved by is reached). mammalian animal models (mice, guinea
using mild to moderate hypothermia ● Speed of rewarming (this should be pigs, rats, etc.). This mechanism plays
(31°C–35°C) rather than deep hypother- slow lest the destructive processes be out within relatively short time frames,
mia (ⱕ30°C), with far fewer and less se- reinitiated; this happens frequently if [⫾2 hrs or less] (25–30). However, clini-
vere side effects. The basis for this new rewarming speeds are high). cal studies in cardiac arrest patients have
insight was the observation that the pro- ● Proper management and prevention of reported significant improvements in
tective effects of hypothermia were not side effects. outcome even when induction of hypo-
(at least not exclusively or mainly) due to thermia was delayed for up to 8 hrs (17);
decreased oxygen and glucose consump- The equation is complicated by the this indicates that other, more protracted
tion in the brain. Indeed, the mecha- fact that the relative contributions of dif- mechanisms must play a role in deter-
nisms involved are far more complex. ferent mechanisms to the ongoing injury mining outcome in human brain injury.
A cascade of destructive events and may differ. There may be differences be- Thus, focusing exclusively on neuroexcito-
processes begins at the cellular level in tween various types of injury (e.g., trau- toxicity would significantly underestimate
the minutes to hours following an initial matic vs. purely postischemic), between the available time window for application of
injury. The injury can be ischemic or different patients (depending on genetic hypothermia.
traumatic. Ischemia may be induced by factors, comorbidities, gender, etc.), and Nevertheless, in spite of such limita-
obstruction of a blood vessel, by pressure/ even within the same patient over time. tions there is much that we can learn
edema, or by other mechanisms. This de- Thus, the available windows of opportu- from animal experiments, in particular if
structive cascade is similar regardless of nity for therapeutic interventions such as a treatment has shown value in more
the cause, although there are differences hypothermia may vary, and the same may than one animal model and/or in pri-
in regard to how long the cascade is sus- apply to the required duration for the mates (orangutans, chimpanzees, etc.).
tained. These processes—which have cooling treatment to be (fully) effective. What we have learned regarding ischemia
been collectively described in terms such For all these reasons, a better under- –reperfusion and posttraumatic injuries,
as postresuscitation disease, reperfusion standing of underlying mechanisms may and the protective effects of hypothermia,
injury, and secondary brain injury—may help us to better target our treatments, is summarized in Figure 1 and are now
continue for hours to many days after the and help improve outcome. discussed in more detail.
initial injury and can be retriggered by Many of the available data come from
new episodes of ischemia (17). animal experiments; translating these re- Underlying Mechanisms,
A key point is that all of these pro- sults into clinical practice poses some Physiological Sequelae, and
cesses are temperature dependent; they limitations. Firstly, the relative impor- Consequences for Patient
are all stimulated by fever, and can all be tance of injury mechanisms may differ Management
mitigated or blocked by mild to moderate between humans and animals, and also
hypothermia. The wide-ranging effects of between different animal species. Thus, if Decrease in Cerebral Metabolism.
hypothermia may explain why it has a specific treatment decreases the degree When hypothermia was first used in a
proved to be clinically effective, whereas of histological brain damage in a specific clinical setting it was presumed that its
studies with agents that affect just one of area in rats, this treatment may have sim- protective effects were due purely to a
the destructive processes have been far ilar histological effects in humans but slowing of cerebral metabolism, leading
less successful (18 –22). may not make a clinically significant dif- to reduced glucose and oxygen consump-
The second important development ference in outcome (23, 24). Secondly, tion. Indeed this assumption is not com-
was the advent of intensive care units some treatments may target specific de- pletely incorrect: Cerebral metabolism
(ICUs) to care for critically ill patients. structive mechanisms playing a central decreases by 6% to 10% for each 1oC

Crit Care Med 2009 Vol. 37, No. 7 (Suppl.) S187


death. Whether apoptosis will develop is
determined by cellular processes such as
mitochondrial dysfunction, other disor-
ders in cellular energy metabolism, and
release of so-called caspase enzymes. Nu-
merous studies have shown that hypo-
thermia can interrupt the apoptotic path-
way, thereby preventing cellular injury
from leading to apoptosis (39 – 43). Hypo-
thermia seems to affect mainly the early
stages of the apoptotic process and apo-
ptosis initiation (40). Its effects include
inhibition of caspase enzyme activation
(39 – 41, 43, 44), prevention of mitochon-
drial dysfunction (42), decreased overload
of excitatory neurotransmitters, and
modification of intracellular ion concen-
trations. (The latter mechanisms are dis-
cussed in more detail below.)
Apoptosis begins relatively late in the
postperfusion and/or posttrauma phase,
while continuing for a period of 48 to 72
hrs or even longer (43– 45). Thus, apo-
Figure 1. Schematic depiction of the mechanisms underlying the protective effects of mild to ptosis is one of the mechanisms that can
moderate hypothermia. TxA2, thromboxane A2. be mitigated (and perhaps even pre-
vented) for some time after injury, pro-
viding (at least in theory) a wide window
reduction in body temperature during This phenomenon does not require treat- of opportunity for therapeutic interven-
cooling (27, 29, 31–35). Early studies at- ment; pH rarely decreases below 7.25, tions such as hypothermia. For this and
tempted to reduce metabolism as far as and the drop in extracellular pH is not other reasons, influencing apoptotic
possible by using deep hypothermia reflected in intracellular pH, which in- pathways could play an important role in
(ⱕ30°C). Our current understanding is creases slightly under hypothermic con- providing neuroprotection and in miti-
that a reduction in metabolic rate is only ditions (33). gating postresuscitation injury in hu-
one of many mechanisms underlying hy- Another consequence of hypothermia mans.
pothermia’s protective effects. is decreased insulin secretion and, in Ion Pumps and Neuroexcitotoxicity. A
Physiological Consequences and Pa- many patients, a moderate (and some- large body of evidence suggests that hy-
tient Management. When core tempera- times severe) insulin resistance. This can pothermia inhibits harmful excitatory
ture drops to 32°C, the metabolic rate lead to hyperglycemia and/or a significant processes occurring in brain cells during
decreases to 50% to 65% of normal; ox- increase in doses of insulin required to ischemia–reperfusion. Levels of high-
ygen consumption and CO2 production maintain glucose levels within an accept- energy metabolites such as adenosine
will decrease by the same percentage. If able range. This temperature-dependence triphosphate and phosphocreatine de-
ventilator settings are left unchanged this of insulin requirements is particularly crease within seconds when oxygen sup-
will lead to hyperventilation, with poten- important in the rewarming phase, when ply to the brain is interrupted (27). The
tially adverse consequences such as cere- insulin sensitivity may increase rapidly, breakdown of adenosine triphosphate and
bral vasoconstriction (36, 37). The de- leading to hypoglycemia if insulin doses the switch of intracellular metabolism to
crease in oxygen consumption will are not decreased. Thus, glucose levels anaerobic glycolysis lead to an increase in
increase oxygen levels in the blood, pos- should be checked frequently particularly intracellular levels of inorganic phosphate,
ing a (theoretical) risk during reperfusion during rewarming and, to a lesser extent, lactate, and H⫹, resulting in both intra- and
because high oxygen levels may increase the induction phase of hypothermia (1). extracellular acidosis and an influx of cal-
reperfusion injury (38). Therefore, venti- For this and other reasons, rewarming cium (Ca2⫹) into the cell. Loss of adenosine
lator settings should be adjusted as tem- rates following hypothermia treatment triphosphate and acidosis also inhibits the
perature decreases, and frequent checks should be slow and controlled. In our mechanisms that normally deal with exces-
of blood gasses are necessary especially in unit the target rewarming rate is sive intracellular Ca2⫹ by sequestering
the induction phase. During mainte- 0.25°C/hr for postcardiac arrest patients Ca2⫹ from the cell, further aggravating in-
nance the feeding rate should also be and 0.1°C/hr for patients following severe tracellular Ca2⫹ overload. These problems
adjusted to reflect lower metabolic de- TBI. are compounded by failure of adenosine
mands. Apoptosis, Calpain-Mediated Proteol- triphosphate– dependent Na⫹-K⫹ pumps
Other metabolic changes include in- ysis and Mitochondrial Dysfunction. Fol- and K⫹, Na⫹, and Ca2⫹ channels, leading
creased fat metabolism, leading to an in- lowing a period of ischemia and reperfu- to an additional influx of Ca2⫹ (46). The
crease in the levels of glycerol, free fatty sion, cells can become necrotic, fully or excess Ca2⫹ induces mitochondrial dys-
acids, ketonic acids, and lactate. This in partially recover, or enter a path leading function (increasing intracellular calcium
turn can cause mild metabolic acidosis. to so-called apoptosis or programmed cell influx yet further, in a vicious cycle) and

S188 Crit Care Med 2009 Vol. 37, No. 7 (Suppl.)


activates numerous intracellular enzyme this may vary between different species duced inflammatory reactions and release
systems (kinases and proteases). In addi- and different types of injury. Some au- of proinflammatory cytokines (75–78).
tion, immediate early genes are activated thors have suggested that the therapeutic Hypothermia also prevents or mitigates
and a depolarization of neuronal cell mem- window could be extended by combining reperfusion-related DNA injury, lipid per-
branes occurs, with a release of large hypothermia with other, more specific oxidation, and leukotriene production
amounts of the excitatory neurotransmitter treatments such as caspase inhibitors or (47– 49), and it decreases the production
glutamate into the extracellular space (25). other experimental compounds (41, 68). of nitric oxide, which is a key agent in the
This leads to prolonged and excessive acti- Immune Response and Inflammation. development of postischemic brain injury
vation of membrane glutamate receptors, In most types of brain injury, a significant (25). In addition, hypothermia can impair
further stimulating Ca2⫹ influx through and protracted inflammatory response be- neutrophil and macrophage function,
activation of Ca2⫹ channels in another vi- gins about 1 hr following a period of isch- and (at temperatures ⬍32°C–33°C) de-
cious cycle. Under normal circumstances, emia–reperfusion. Proinflammatory medi- crease white blood cell count.
neurons are exposed to only very brief ators such as tumor necrosis factor-␣ and In animal experiments, the extent of
pulses of glutamate; prolonged glutamate interleukin-1 are released in large quanti- brain injury and infarct size can be sig-
exposure induces a permanent state of hy- ties by astrocytes, microglia, and endothe- nificantly attenuated if any of these pro-
perexcitability in the neurons (the excito- lial cells; this rise begins ⫾1 hr after injury cesses is mitigated or interrupted (25);
toxic cascade), which can lead to additional and they remain elevated for up to 5 days given that hypothermia can affect all of
injury and cell death. Furthermore, high (27, 69). This stimulates the chemotaxis of these steps there is (at least in theory) a
levels of glutamate can be neurotoxic, es- activated leukocytes across the blood– brain huge potential for improving outcome.
pecially in energy-deprived cells. Glutamate barrier, leading to an accumulation of in- Furthermore, given that the inflamma-
receptor activation can persist for some flammatory cells in the injured brain, as tory response begins relatively late (⬎1
time after reperfusion, even when gluta- well as the appearance of adhesion mole- hr following ischemia–reperfusion), and
mate levels have returned to normal; this cules on leukocytes and endothelial cells. given that the destructive processes take
may be another important mediator of Simultaneously, there is an activation of some time to develop fully and continue
brain cell death. the complement system which begins in for prolonged periods of time, there ap-
In summary, ischemia and reperfusion the very early stages after brain injury and pears to be a clear therapeutic window for
lead to an interruption of a delicate bal- further stimulates the passage of neutro- use of hypothermia to favorably affect
ance between calcium influx and seques- phils and (in later stages) monocytes- this specific mechanism.
tration at the cellular level. Numerous macrophages (69). These inflammatory and Free Radical Production. A destruc-
animal experiments have clearly demon- immunologic responses occur especially tive process that is closely linked to but
strated that key destructive processes of during reperfusion and are accompanied by distinct from the mechanisms discussed
the neuroexcitatory cascade (such as cal- free radical production. This can cause sig- above is the release of free oxygen radi-
cium influx, accumulation of glutamate, nificant (additional) injury through the cals following ischemia–reperfusion. Me-
and the release of its coagonist glycine) phagocytic actions of macrophages, synthe- diators such as superoxide (O2⫺), per-
can be prevented, interrupted, or miti- sis of toxic products, and further stimula- oxynitrite (NO2⫺), hydrogen peroxide (H2
gated by hypothermia (23, 25, 45–53). tion of immune reactions in a vicious cycle. O2), and hydroxyl radicals (OH⫺) play an
Even a relatively small decrease in tem- However, it should be realized that important role in determining whether
perature can significantly improve ion this inflammatory response is to some injured cells will recover or die (47, 50,
homeostasis, whereas the occurrence of extent physiological, and may have a dual 79, 80). Free radicals can oxidize and
fever can trigger and stimulate these de- role in the sense that some inflammatory damage numerous cellular components.
structive processes. mediators have neuroprotective proper- Although brain cells have various enzy-
It is unclear how long the window of ties, whereas others are neurotoxic (69 – matic and nonenzymatic antioxidant
opportunity to interrupt this cascade is. 72). Nevertheless, there is strong evi- mechanisms that prevent this type of in-
Disruptions in Ca2⫹ homeostasis begin in dence suggesting that certainly a jury under normal circumstances, free
the minutes after injury but may con- disproportionate and persistent produc- radical production following ischemia–
tinue for many hours (sometimes even tion of cytokines and leukocyte infiltra- reperfusion is so great that these defen-
days). Furthermore, the mechanism can tion can significantly increase the risk sive mechanisms are likely to be over-
be reinitiated by new episodes of isch- and extent of brain cell injury and infarc- whelmed, leading to peroxidation of
emia. Thus, in theory this mechanism tion (68 –74). Especially the interleukin-1 lipids, proteins, and nucleic acids.
may be susceptible to therapeutic inter- family appears to be important in this Under hypothermic conditions the
ventions provided these are initiated in regard (74). Of note, this effect is to some quantity of free radicals that is generated
the hours following injury. However, extent time dependent, with the destruc- is significantly reduced, although free
some animal experiments suggest that tive aspects of inflammation outweighing radical production is not completely pre-
neuroexcitotoxicity can be blocked or re- the potential benefits especially in the vented (47, 50, 79, 80). This allows the
versed only if the treatment is initiated in later stages of injury (69 –72). Thus, endogenous antioxidative (protective)
the very early stages of the neuroexcita- again, there may be a potential time win- mechanisms to better cope with free rad-
tory cascade (26 –27, 30, 54). Other stud- dow for therapeutic interventions to in- icals that are being released, thereby pre-
ies have reported somewhat wider time terrupt or mitigate this process before it venting or significantly mitigating oxida-
frames, ranging from 30 mins to up to 6 becomes destructive. tive damage. This allows the cell to repair
hrs (55– 67). Thus, the period during Numerous animal experiments and itself and recover, rather than suffering
which the neuroexcitatory cascade can be some clinical studies have shown that permanent damage and/or dying. The de-
modified in vivo remains unclear; indeed, hypothermia suppresses ischemia-in- gree of inhibition of free radical produc-

Crit Care Med 2009 Vol. 37, No. 7 (Suppl.) S189


window to mitigate neurological injuries.
Brain edema may develop rapidly, but
more typically arises several hours after
injury, often peaking after 24 to 72 hrs
(94). It should be noted that although
hypothermia has been shown to decrease
ICP in many clinical studies, the results
in improving survival and neurological
outcomes have been mixed (17, 88), with
one large trial failing to show any benefits
during hypothermia treatment in spite of
hypothermia-induced decreases in ICP
(95). These varying results may be
(partly) linked to the management of hy-
pothermia’s side effects (17, 88); these
issues are discussed more extensively be-
low. As explained above, the speed of in-
duction, duration and depth of hypother-
mia, and rewarming speeds all play
important roles in determining whether
or not lasting protective effects are
Figure 2. Schematic depiction of the potential role of intracranial pressure as both a marker of achieved (1, 17, 88, 96, 97).
ongoing brain injury and a potential cause of additional injury, and of the factors that may contribute
to a rise in intracranial pressure. CSF, cerebrospinal fluid. Intra- and Extracellular Acidosis and
Cellular Metabolism. The diminished in-
tegrity of cell membranes, the failure of
tion appears to be more or less linearly scribed above (dampening inflammatory various ion pumps, development of mito-
linked to the temperature; i.e., the lower responses and free radical production, chondrial dysfunction, inappropriate ac-
the temperature, the lower the amount of improving ion homeostasis, etc.), and tivation of numerous enzyme systems
free radicals (79). through other mechanisms, which are with cellular hyperactivity, and the dis-
Vascular Permeability, Blood–Brain summarized in Figure 2. Brain edema ruption of various other intracellular pro-
Barrier Disruption, and Edema Forma- can be monitored by measuring intracra- cesses all contribute to the development
tion. Ischemia–reperfusion and/or trau- nial pressure (ICP), which can be re- of intracellular acidosis, a factor that
matic injury can lead to significant dis- garded as a final common pathway for the powerfully stimulates many of the above-
ruptions in the blood– brain barrier, destructive processes leading to brain mentioned destructive processes (98, 99).
which can facilitate the subsequent devel- edema (88) [Fig. 2]. Ischemia–reperfusion also leads to sub-
opment of brain edema (81– 83). Thera- The key role of brain edema and intra- stantial rises in cerebral lactate levels
peutic interventions such as mannitol ad- cranial hypertension as a cause of (addi- (100 –102). All of these factors can be
ministration in TBI or stroke can add to tional) neurological injury in severe TBI significantly attenuated by hypothermia
blood– brain barrier disruption (82). Mild and ischemic stroke is well recognized (98 –103).
hypothermia significantly reduces blood– (89, 90). Brain edema can also increase In addition, brain glucose utilization
brain barrier disruptions (81– 83), and brain injury in acute encephalitis and is affected by ischemia–reperfusion, and
also decreases vascular permeability fol- meningitis (91), and some evidence sug- there is evidence suggesting that hypo-
lowing ischemia–reperfusion, further de- gests that edema-related injury can play a thermia can improve brain glucose me-
creasing edema formation (84). The con- role in some patients with posthypoxic tabolism; i.e., the ability of the brain to
cept of a membrane- and blood– brain injury following cardiac arrest (92). utilize glucose (104, 105). Similar obser-
barrier–stabilizing effect of hypothermia As intracranial hypertension appears vations have been made in severe TBI, for
is supported by the observation that hy- to be both a marker for ongoing neuro- which animal experiments and some clin-
pothermia decreases extravasation of he- logical injury and a cause of additional ical studies suggest that an initial in-
moglobin following TBI (85). Ischemia– injury (88), it seems plausible that treat- crease in cerebral glucose metabolism in
reperfusion affects blood– brain barrier ments that reduce ICP will also help im- the hours following trauma is followed by
integrity in other ways, including de- prove neurological outcome. ICP is fre- a deep and persistent decrease in meta-
creased fluidity and integrity of cell mem- quently used as a parameter to guide bolic rate, with a depression of mitochon-
branes and increased vascular permeabil- treatments and assess short-term thera- drial oxidative phosphorylation and glu-
ity of microvascular endothelial cells in peutic efficacy in patients with TBI (89, cose utilization that can last for several
the brain, mediated by vascular endothe- 93). Hypothermia has been used to treat weeks (106 –108). Induced hypothermia
lial growth factor via release of nitric brain edema and reduce ICP in a wide applied during or after reperfusion in-
oxide (86, 87). range of neurological injuries, including creases the speed of metabolic recovery,
These processes of membrane disinte- TBI, ischemic stroke, hepatic encepha- with a better preservation of high-energy
gration and hypoxia-induced leakage can lopathy, meningitis, encephalitis, and phosphates and reduced accumulation of
be mitigated or reversed by hypothermia subarachnoid hemorrhage (17, 28). toxic metabolites (106 –108).
(87). Hypothermia also decreases cyto- In theory, this mechanism of action Brain Temperature and Cerebral
toxic edema via the mechanisms de- would offer a relatively wide therapeutic Thermopooling. Even in healthy individ-

S190 Crit Care Med 2009 Vol. 37, No. 7 (Suppl.)


uals, the temperature of the brain is (1°C–2°C above normal for 3 hrs) was Hypothermia has some anticoagula-
slightly higher than the measured core induced 24 hrs after a brief episode of tory effects. Mild platelet dysfunction oc-
temperature (109 –111). This difference forebrain ischemia. This suggests that fe- curs at temperatures ⱕ35°C, and some
can increase significantly in patients fol- ver can be detrimental even when it is of inhibition of the coagulation cascade de-
lowing neurological injury, with gradi- short duration and occurs on the day velops at temperatures ⱕ33°C; platelet
ents ranging from 0.1°C to more than after injury. Others have reported similar count can also decrease during cooling
2°C (112–118), although this phenome- observations in different animal models (1, 151–154). In theory, this anticoagula-
non does not occur in all brain-injured (132–136). The effects of fever appear to tion effect may constitute yet another
patients (118). These differences may in- be more pronounced when they coincide neuroprotective mechanism. This re-
crease even further when a patient devel- with an episode of cerebral ischemia, sug- mains speculative given that no studies
ops fever, a very frequent phenomenon in gesting that ischemic brain cells become directly addressing this issue have been
patients with neurological injuries (119 – even more susceptible to the harmful ef- performed.
123). In addition, there may be small fects of hyperthermia (135–136). Vasoactive Mediators. Several studies
temperature differences between differ- Numerous clinical studies have con- have shown that hypothermia affects the
ent areas of the brain even in healthy firmed that fever is indeed an indepen- local secretion of vasoactive substances
individuals, with active areas having a dent predictor of adverse outcome in such as endothelin, thromboxane A2
slightly higher temperature (124 –127). stroke, TBI, and postanoxic injury follow- (TxA2), and prostaglandin I2 in the brain
These differences increase significantly ing cardiac arrest (121–123, 137–142). and at other sites. Endothelin and TxA2
when brain injury occurs, with injured Schwarz et al (121) found that even very are powerful vasoconstrictive agents,
areas becoming more hyperthermic than mild hyperthermia (⬎37.5°C!) within the whereas prostaglandin I2 is a vasodilator.
noninjured areas (128). The reason for first 72 hrs was independently associated TxA2 also stimulates platelet aggregation
this is that some of the destructive mech- with poor outcome in patients with intra- (155–157). TxA2 and prostaglandin I2 play
anisms discussed above lead to excessive cerebral hemorrhage, and that there was an important role in regulating local ce-
generation of heat. These include excito- a linear relationship between the severity rebral blood flow, and a balance between
toxicity (which, as described above, in- and duration of fever and the risk for the two is required to maintain ho-
duces “cellular hyperactivity” with ex- adverse outcome. In patients with isch- meostasis (155). This local homeostasis
emic stroke, fever is associated with a can be disrupted following an ischemic or
treme activation of enzyme systems, with
3.4-fold increase in risk for adverse out- traumatic event, with a relative increase
concomitant generation of heat), the
come (140), a higher brain infarct volume in production of TxA2 (158 –161). This
neuroinflammatory response, free radical
(141), and increased mortality (142). Al- disruption in equilibrium can lead to va-
production, and a process known as “ce-
though it has not been conclusively proven soconstriction, hypoperfusion, and
rebro thermopooling” (128, 129), mean-
that these relationships are causal (i.e., that thrombogenesis in injured areas of the
ing that excess heat generated in injured
fever increases injury rather than just being brain.
areas of the brain becomes more difficult
a marker), this is strongly suggested by the Animal experiments and small clinical
to remove via the normal heat dissipation
temporal relationship and the results from studies suggest that this relative predom-
mechanisms (lymph and venous drain- animal studies (130 –136). inance of local vasoconstrictors can be
age) due to development of local brain Coagulation Activation and Forma- corrected or modified by hypothermia
edema. The heat thus becomes trapped in tion of Microthrombi. Cardiopulmonary (49, 162, 163). Aibiki et al (162) reported
injured areas, further adding to hyper- arrest and resuscitation are accompanied that moderate hypothermia (32°C–33°C)
thermia-related injury. by a marked activation of coagulation, led to a reduction in prostanoid produc-
Thus, a vicious cycle can arise: Brain which can lead to intravascular fibrin for- tion and attenuation of the imbalance
injury leads to general overheating of the mation with blockage of the microcircu- between TxA2 and prostaglandin I2 in pa-
brain; more overheating occurs especially lation in the brain and heart (143–146). tients with severe TBI. Chen et al (163)
in injured areas; this leads to local and Administration of anticoagulants such as found decreased production of the vaso-
sometimes general brain edema, which heparin and recombinant tissue-type constrictor endothelin-1 during induc-
then makes it more difficult to remove plasminogen activator improves micro- tion of hypothermia.
the excess heat. Due to this chain of circulatory reperfusion and survival in This and other preliminary evidence
events, brain temperatures in injured animal experiments (146, 147); in addi- suggests that hypothermia may favorably
brain areas may exceed measured core tion, thrombolysis can improve cerebral affect vasoactive mediator secretion in
temperatures by as much as 2°C to 4°C. tolerance to ischemia (148). Initial clini- brain-injured patients. However, regula-
This is relevant because there is cal reports suggesting that administra- tion of cerebral perfusion is a highly com-
strong evidence from animal studies that tion of thrombolytic agents could im- plex issue, especially in the injured brain;
(external) induction of hyperthermia sig- prove neurological outcome and survival local circulation can be influenced by the
nificantly increases the risk and extent of in cardiac arrest (149) have not been con- presence or absence of cerebral autoreg-
neurological injury (17, 130 –136). Hy- firmed in larger studies (150). Neverthe- ulation, ventilator settings and blood gas
perthermia increases the likelihood that less, activation of coagulation seems to management, systemic blood pressure,
ischemic areas become apoptotic or ne- play an important role in developing isch- fluid management including osmotic
crotic, even when fever develops (or is emia–reperfusion injury, and although therapy, etc. Thus, the impact of hypo-
externally induced) some time after the reversing this mechanism by itself may thermia on this equation needs to be
initial injury. Baena et al (131) reported a not improve outcome, this may be differ- studied in greater detail.
2.6-fold increase in neuronal injury in ent if additional mechanisms are targeted Improved Tolerance for Ischemia. Hy-
the rat hippocampus if mild warming simultaneously. pothermia improves the tolerance for

Crit Care Med 2009 Vol. 37, No. 7 (Suppl.) S191


depressionlike depolarizations (23, 179).
This has been studied mainly in models
for TBI and stroke; its potential role in
the relatively brief global ischemic brain
injury that characterizes cardiac arrest is
unclear. Hypothermia can suppress
spreading depressions in different types
of neurological injury (23, 180 –182), pro-
viding yet another mechanism by which
hypothermia could improve outcome.
Influence on Genetic Expression. Hy-
pothermia increases the expression of so-
called immediate early genes, which are
a part of the protective cellular stress
response to injury, and stimulates the
induction of cold shock proteins, which
can protect the cell from ischemic and
Figure 3. Graphic depiction of the three phases of hypothermia treatment. The induction phase traumatic injury (97). This provides yet
should last between 30 and 120 mins; rapid cooling may lead to a small overshoot, which should be another mechanism through which hy-
accepted provided it is no greater than 1°C. The maintenance phase usually lasts 24 hrs in cardiac pothermia can provide neuroprotection.
arrest patients (may be longer for other indications) and should be characterized by no or minimal
fluctuations in temperature. The rewarming phase should be slow and controlled, with rewarming Thus, hypothermia favorably affects a
rates of 0.2°C to 0.5°C in cardiac arrest patients and lower rewarming rates for other indications. Fever whole range of destructive mechanisms
should be prevented after rewarming. unfolding in the injured brain after isch-
emia or trauma. This wide range of ef-
fects probably explains its efficacy, in
ischemia in various animal models (164 – patients with postanoxic encephalopathy, contrast to interventions that target just
166). For this reason, it is widely used in stroke, TBI, and subarachnoid hemor- one or two of these mechanisms and have
the perioperative setting, especially in rhage (168 –172). It remains uncertain mostly proved unsuccessful in clinical
major vascular surgery, cardiothoracic whether nonconvulsive status is a direct trials. Different mechanisms may play a
surgery, and neurosurgical interventions cause of additional injury; however, greater or smaller role in different types
(17). An ability to better withstand peri- mounting evidence suggests that brain of brain injury, and/or during different
ods of ischemia would be an important injury can be significantly increased phases of ongoing brain injury. A more
protective mechanism, because many when nonconvulsive status occurs in the detailed understanding of these processes
types of neurological injury can be com- acute phase of brain injury; i.e., while the will help us apply therapeutic hypother-
plicated by ischemic events in the days destructive processes outlined above are mia and controlled normothermia more
following the initial insult. ongoing (168, 169). In other words, al- effectively.
For example, in patients with TBI a though the role of nonconvulsive status The other key element for successful
combination of biological factors (cyto- by itself has not been fully clarified, when use of therapeutic cooling is awareness
toxic and vasogenic edema) and mechan- it occurs in combination with ongoing and proper management of the physio-
ical factors (blockage of spinal fluid brain injury the two are synergistically logical consequences and side effects.
drainage) can lead to cerebral edema and detrimental (168, 169). These issues are dealt with in the next
intracranial hypertension in the hours Evidence from various sources shows section.
and days following initial injury (Fig. 2); that hypothermia can suppress epileptic
cerebral edema can then cause additional activity. Small case series report success-
Physiological Aspects of
ischemic injury. In patients with sub- ful use of cooling to treat grand mal sei-
arachnoid hemorrhage the development zures (173–175). Animal studies have
Cooling
of vasospasms in the days and weeks fol- shown that external warming increases Phases of Cooling Treatment. A hypo-
lowing a bleeding episode can lead to the extent of epilepsy-induced brain in- thermia treatment cycle can be divided
significant ischemic injury. The risk for jury, that prevention or prompt treat- into three distinct phases (Fig. 3):
ischemic injury occurring in the postad- ment of fever reduces these injuries, and
mission period may also apply to postcar- that induction of hypothermia further de- ● The induction phase, when the aim is
diac arrest patients, in whom cerebral creases seizure-induced brain injury to get the temperature below 34°C and
ischemia may persist for several hours (176 –179). This antiepileptic effect offers then down to the target temperature as
following successful resuscitation even yet another pathway through which hy- quickly as possible;
when arterial oxygen levels are normal pothermia could provide neuroprotec- ● The maintenance phase, when the aim
(167). Thus, a hypothermia-induced in- tion, and by which fever may increase is to tightly control core temperature,
crease in tolerance for ischemia could be neurological injury. with minor or no fluctuations (maxi-
another powerful protective mechanism. Spreading Depressionlike Depolariza- mum, 0.2°C– 0.5°C);
Suppression of Epileptic Activity. tions. Some animal studies have sug- ● The rewarming phase, with slow and
Nonconvulsive status epilepticus (i.e., ep- gested that neuronal damage in various controlled warming (target rate,
ileptic activity without obvious clinical types of neurological injury can be signif- 0.2°C– 0.5°C/hr for cardiac arrest pa-
signs and symptoms) occurs frequently in icantly increased by so-called spreading tients and 0.1°C– 0.2°C/hr for other

S192 Crit Care Med 2009 Vol. 37, No. 7 (Suppl.)


categories such as patients with severe sured at other sites remained normal. Al- sodilation, which can facilitate heat loss
TBI) (183). though none of these experiments directly through surface cooling.
addressed optimal rewarming rates for car- Relatively effective and rapidly acting
Each of these phases has specific man- diac arrest patients treated with hypother- antishivering agents include fentanyl, al-
agement problems. The risk for immedi- mia, these and other findings indicate that fentanyl, meperidine, dexmedetomidine,
ate side effects such as hypovolemia, elec- rapid rewarming can increase ischemia and propofol, clonidine, and magnesium (1).
trolyte disorders, and hyperglycemia is aggravate destructive processes in the in- Specific advantages and disadvantages of
greatest in the induction phase (17, 184, jured brain (1). these and other agents are reviewed in
185). This phase presents the greatest Another important concept is the more detail elsewhere (1). Brief-acting
patient management problems, requiring maintenance of strict normothermia fol- paralyzing agents can be useful in the
frequent adjustments in ventilator set- lowing the rewarming phase. As dis- induction phase, especially when hypo-
tings; dosing of sedation, insulin, and va- cussed above, fever is independently thermia is initiated outside the ICU set-
soactive drugs; and fluid and electrolyte linked to adverse outcome in all types of ting, but prolonged paralysis is usually
administration (185). The risks can be neurological injury, including postanoxic unnecessary (1). The effectiveness of the
reduced by rapid induction of hypother- injury following cardiac arrest (17). In shivering response decreases with age;
mia; i.e., minimizing the duration of the addition, cerebrovascular reactivity may therefore, the doses of drugs required to
induction phase and reaching the more be impaired following hypothermia treat- suppress shivering are usually lower in
stable maintenance phase as quickly as ment (192), increasing the potential older patients. Warm-air skin counter-
possible. Rapid cooling can be achieved harmful effects of fever. warming can be used as an accessory
by combining different cooling methods, method to lower the shivering threshold,
for example, surface cooling with infu- and to decrease drug doses required to
sion of 1500 –3000 mL of cold (4°C) fluids Physiological Changes and Side
prevent shivering (198, 199).
using a pressure bag (186). Furthermore, Effects of Cooling
In our unit, the standard approach is
some of the new intravascular devices to administer a loading dose of magne-
may have more rapid cooling rates than The induction of mild hypothermia in-
duces numerous changes throughout the sium (30 mmol) and fentanyl (50 –100
previously used methods (1). ␮g) when cooling is initiated, together
The maintenance phase is characterized body. The most important physiological
changes and side effects, and their conse- with continuous infusion of either mida-
by increased stability of the patient, with a
quences for patient management, are dis- zolam or propofol (the latter only in he-
decreased shivering response and less risk
cussed below. modynamically stable patients). If shiver-
for hypovolemia and electrolyte loss. In this
Shivering and Cutaneous Vasoconstric- ing occurs, another bolus of fentanyl
phase, attention should shift toward pre-
tion. A separate article in this supplement (50 –150 ␮g) is given, and more magne-
vention of longer-term side effects such as
is devoted to shivering (193); therefore, sium if serum Mg2⫹ is ⱕ2.0 mmol/L. In
nosocomial infections and bedsores.
this topic is discussed only briefly here. some patients, the dose of sedatives will
In the rewarming phase, the patient’s
In awake patients, shivering induces be temporarily increased during induc-
temperature should be increased very
slowly, for a number of reasons. Firstly, unfavorable effects such as increased ox- tion and/or a bolus dose of midazolam
rapid rewarming can cause electrolyte dis- ygen consumption and metabolic rate, (5–10 mg) will be given. If shivering per-
orders (in particular, hyperkalemia) caused excess work of breathing, and increased sists, one of the following drugs is given,
by shifts from the intracellular to the ex- heart rate with increased myocardial ox- depending on the clinical circumstances:
tracellular compartment. This can be ygen consumption (1, 184, 194 –197). In clonidine, meperidine, ketanserin, or (in
largely prevented by slow and controlled the perioperative setting, hypothermia rare cases) a brief-acting paralyzing
rewarming. Secondly, insulin sensitivity has been linked to an increased risk for agent. All patients should be carefully
can increase during rewarming, and slow morbid cardiac events, particularly in monitored for shivering during all phases
rewarming decreases the risk for hypogly- older patients with heart disease (194 – of hypothermia treatment. Use of a re-
cemia if the patient is being treated with 196). However, these adverse conse- cently developed shivering assessment
insulin. Thirdly, some animal experiments quences are linked to the hemodynamic scale (200) may be considered for this
and clinical observations suggest that rapid and respiratory responses rather than to purpose.
rewarming could lead to loss of some or shivering per se; these responses can be The importance of adequate sedation
even all of the protective effects of hypo- largely suppressed through administra- and suppression of hypothermia-induced
thermia (187–191). Significant decreases in tion of sedatives, anesthetics, opiates, or stress responses is underscored by obser-
jugular venous oxygen saturation have other drugs during therapeutic cooling vations from animal experiments sug-
been reported during rapid rewarming of (1). For example, in contrast to the tachy- gesting that some or all of hypothermia’s
patients following cardiac surgery under cardia associated with perioperative (ac- neuroprotective effects can be lost if cool-
hypothermic conditions, indicating hyp- cidental) hypothermia, heart rates are ing is used in nonsedated animals. One
oxia of the brain (190); more slow rewarm- markedly reduced during therapeutic research group observed no protective ef-
ing led to a decrease in the incidence and cooling (1). Nevertheless, it is important fects when cooling was used after global
severity of jugular bulb desaturations (190). to prevent or aggressively treat shivering anoxia in unsedated newborn piglets
In another study, Bissonnette et al (191) because it significantly complicates hypo- (201); when the experiment was repeated
reported that patients who were rapidly re- thermia induction, and leads to an unde- in the same model under adequate seda-
warmed following cardiopulmonary bypass sirable increase in metabolic rate and ox- tion, significant improvements in out-
surgery often developed severe brain hyper- ygen consumption (1). Appropriate come were noted in hypothermia-treated
thermia, even when core temperature mea- sedation and analgesia will also cause va- animals (202). The authors concluded

Crit Care Med 2009 Vol. 37, No. 7 (Suppl.) S193


that stress effects of cooling in unsedated In a sample assayed at 37°C: be kept in the high-normal range during
animals could negate protective effects. and after hypothermia treatment.
Although this phenomenon has not been ● Subtract 5 mm Hg PO2 per 1°C that the Extra care should be taken during re-
well studied in humans, it seems reason- patient’s temperature is ⬍37°C; warming because hyperkalemia may de-
able to avoid a cooling-related stress re- ● Subtract 2 mm Hg PCO2 per 1°C that velop during this phase, due to release of
sponse. the patient’s temperature is ⬍37°C; potassium sequestered to the intracellular
Metabolism, Blood Gases, Glucose ● Add 0.012 pH units per 1°C that the compartment during hypothermia induc-
and Electrolytes. Hypothermia decreases patient’s temperature is ⬍37°C. tion (1). Hyperkalemia can be prevented by
the metabolic rate by ⫾8% per 1oC drop slow and controlled rewarming, allowing
in core temperature, with a parallel de- Thus, a sample from a patient with a the kidneys to excrete the excess potas-
crease in oxygen consumption and pro- core temperature of 33°C analyzed in the sium. Thus, the risk for rebound hyperka-
duction of carbon dioxide. This means lab at 37°C with the results pH 7.45, PCO2 lemia in the rewarming phase should not
that ventilator settings need to be ad- 35 mm Hg, and PO2 80 mm Hg would be regarded as a reason to accept hypoka-
justed frequently especially in the induc- have the following temperature-corrected lemia in the induction and maintenance
tion phase of hypothermia, to avoid acci- values: pH 7.50, PCO2 27 mm Hg, and PO2 phase. However, in patients with anuria or
dental hyperventilation that can cause 60 mm Hg. severe oliguria, renal replacement therapy
cerebral vasoconstriction. Blood gas val- In our unit, we use temperature- should be started before the patient is re-
ues are temperature dependent, and if corrected blood gas values but avoid se- warmed.
blood samples are warmed to 37°C before vere hypercapnia. Target PCO2 values at Cardiovascular and Hemodynamic
analysis (as is common in most laborato- 32°C (our usual target temperature in Effects. Hypothermia has complex and
ries), PO2 and PCO2 will be overestimated cardiac arrest patients) are 32 to 36 mm opposing effects on the myocardium and
and pH underestimated in hypothermic Hg, which would be 42 to 46 mm Hg in myocardial contractility, depending on
patients (1). This topic has been reviewed an uncorrected sample. the patient’s volume status and whether
elsewhere (203). Hypothermia can also decrease insulin or not the patient is adequately sedated.
Blood gas management when CO2 val- sensitivity and the amounts of insulin Under normal circumstances, mild hypo-
ues measured at 37°C are kept constant secreted by the pancreas. This can lead to thermia will decrease heart rate and in-
(for example, at 40 mm Hg) regardless of hyperglycemia and/or an increase in the crease myocardial contractility in sedated
the patient’s actual core temperature is doses of insulin required to maintain glu- and euvolemic patients (1, 207–211). Sys-
called alpha-stat management. The alter- cose levels within target range. Sustained tolic function will improve but a mild
native is pH-stat management, in which hyperglycemia has been linked to adverse degree of diastolic dysfunction may occur
PCO2 is corrected for temperature and is outcome in critically ill patients, and may in some patients (1, 207–211). Blood
maintained at a prespecified value. This pose additional risks in patients with neu- pressure remains stable or increases
implies that the temperature-corrected rological injuries (39), although the opti- slightly in most patients during mild
pH will remain constant during pH-stat mal targets for glucose control remain hypothermia (1, 186, 211–213). Cardiac
management and will increase during al- controversial (39). Prevention and/or output decreases along with the heart
pha-stat management (1). Which of these prompt correction of severe hyperglyce- rate; however, the hypothermia-in-
two methods is superior remains to be mia should be part of the therapeutic duced decrease in metabolic rate usu-
determined, and may depend in part on strategy during hypothermia treatment. ally equals or exceeds the decrease in
whether or not cerebral autoregulation is Furthermore, it should be realized that cardiac output, so that the balance be-
preserved (1). Strictly applied alpha-stat doses of insulin required to maintain nor- tween supply and demand remains con-
management could lead to hyperventila- moglycemia are likely to decrease when stant or improves (1).
tion and cerebral vasoconstriction; strict the patient is rewarmed; this means that The heart rate can be artificially in-
pH-stat management could lead to hyper- hypoglycemia can easily develop in the creased by external pacing, or through
capnia, cerebral vasodilation, and in- rewarming phase as insulin sensitivity is administration of chronotropic drugs
creased ICP. Which option is preferable restored, particularly if the patient is re- such as isoprenalin or dopamine. How-
may depend on the precise nature of the warmed (too) quickly. ever, this is usually unnecessary; further-
neurological injury and the presence or Cooling can also affect electrolyte lev- more, myocardial contractility may be
absence of brain edema. Excessive hypo- els. A combination of hypothermia- adversely affected (210, 211, 214). Two
capnia can increase ischemia in injured induced intracellular shift and tubular animal studies (210, 214) and one clinical
areas of the brain, while excessive hy- dysfunction leading to an increase in re- study in patients undergoing cardiac sur-
percapnia can increase brain edema; nal excretion of electrolytes can lead to gery (211) reported that while increasing
thus both can cause additional brain depletion of magnesium, potassium, and heart rate under normothermic conditions
injury. These issues are more exten- phosphate during cooling (185). These increased myocardial contractility and car-
sively addressed in a recently published electrolyte disorders can increase the risk diac output, increasing the heart rate under
review (1). for arrhythmias and other potentially hypothermic conditions significantly de-
For accurate temperature correction, harmful complications (204, 205). There creased myocardial contractility. When
blood samples should be analyzed at the is evidence suggesting that magnesium heart rates were not artificially increased,
patient’s real temperature; this can be could play a role in mitigating various induction of mild hypothermia improved
most easily accomplished by performing types of brain injury (204, 205); hy- myocardial contractility (211).
on-site analysis in the ICU. If this is not pophosphatemia has been linked to respi- Thus, in most patients the heart rate
possible, the blood gas values can be es- ratory problems and increased infection should be allowed to decrease along with
timated in the following way. risk (206). Thus, electrolyte levels should the patient’s core temperature; further-

S194 Crit Care Med 2009 Vol. 37, No. 7 (Suppl.)


more, it should be realized that normal val- shock should not be regarded as a coun- tachycardia) in young infants (233–235).
ues for heart rate change with temperature. terindication for hypothermia treatment. All of this indicates that mild hypother-
Despite these data, hypothermia is of- Hypothermia also causes changes in mia decreases rather than increases the
ten still viewed as a (potential) cause of the heart rhythm and electrocardiogram. risk for arrhythmias, and facilitates
hypotension and (additional) myocardial The hypothermia-induced increase in ve- rather than complicates the treatment of
dysfunction. There are several reasons for nous return discussed above initially arrhythmias.
this: leads to mild sinus tachycardia. Tachy- However, this changes if core temper-
Firstly, in contrast to mild hypother- cardia can be much more pronounced if ature drops below 28°C (in rare cases,
mia, deep hypothermia (⬍30°C) does de- the patient is insufficiently sedated, and ⬍30°C in patients with severe electrolyte
crease myocardial contractility (1, 215). the shivering response with increased ox- disorders and/or severe myocardial isch-
Secondly, hypothermia can cause hypo- ygen consumption and heat generation emia). More profound hypothermia does
volemia by inducing “cold diuresis” can lead to more marked tachycardia. increase the risk for arrhythmias, usually
through a combination of increased ve- This is soon followed by sinus brady- beginning with atrial fibrillation, which
nous return, activation of atrial natri- cardia when core temperature drops be- can progress to more severe arrhythmias
uretic peptide, decreased levels of antidi- low 35.5°C. The heart rate decreases pro- including ventricular tachycardia and
uretic hormone and renal antidiuretic gressively as core temperature drops, and ventricular fibrillation if temperature de-
hormone receptor levels, and tubular at 33°C the normal heart rate will be 45 creases further. Thus, a de novo develop-
dysfunction (185, 216 –221). The increase to 55 bpm (although there is wide inter- ment of atrial fibrillation in a patient with
in venous return is caused by constric- patient variability). The underlying a core temperature ⬍30°C should be
tion of peripheral vessels (particularly in mechanism is a decrease in the rate of viewed as a warning sign, and the patient
the skin) due to hypothermia-induced in- spontaneous depolarization of cardiac should be rewarmed to ⱖ30°C immedi-
creases in plasma norepinephrine levels pacemaker cells (including those of the ately. In general, a temperature drop be-
and activation of the sympathetic nerve sinus node), as well as prolongation of low 30°C should be strenuously avoided
system. This leads to a shift of the blood the duration of action potentials and a during therapeutic cooling in the ICU,
from peripheral (small) veins to deeper mild decrease in the speed of myocardial but cooling treatment should not be
veins and the core compartment of the impulse conduction. The most common withheld because of the presence or risk
electrocardiogram changes are prolonged for arrhythmias.
body, increasing the venous return. In
PR intervals, increased QT interval, and In addition, attending physicians and
this way, hypothermia can induce hypo-
widening of the QRS complex. Some- nurses should be aware that the myocar-
volemia, which if uncorrected can cause
times so-called Osborne waves can be dium becomes more sensitive to mechan-
hypotension as well as electrolyte deple-
seen, although this is relatively rare dur- ical manipulations during deep hypother-
tion and an increase in blood viscosity.
ing mild hypothermia. mia. For example, if a physician decides
The risk for hypovolemia increases signif-
These electrocardiogram changes usu- to perform chest compressions because of
icantly if the patient is simultaneously
ally do not require treatment. It is often severe bradycardia at a core temperature
treated with diuretic agents such as man-
assumed that mild hypothermia increases ⬍28°C, this manipulation can induce ar-
nitol. However, hypotension can be quite the risk for arrhythmias and decreases rhythmias including ventricular fibrilla-
easily prevented by avoiding or promptly the chance of successful treatment of ar- tion (1). An additional problem is that if
correcting hypovolemia, and by avoiding rhythmias, because the hypothermic arrhythmias do develop during deep hy-
excessive stimulation of the heart rate. myocardium becomes less responsive to pothermia, they become significantly
Three studies in pediatric patients en- antiarrhythmic drugs and more difficult more difficult to treat, because the hypo-
rolling a total of 127 patients and two to defibrillate. However, in general this thermic myocardium can become less re-
small studies in adult patients enrolling applies only to deep hypothermia sponsive to antiarrhythmic drugs (1).
18 patients have reported successful use (ⱕ28°C); experimental evidence suggests Furthermore, other treatments for atrial
of mild hypothermia (32°C–33°C) as res- that mild hypothermia can actually de- fibrillation, such as electric cardiover-
cue therapy to reverse refractory cardiac crease the risk for arrhythmias, by stabi- sion, are frequently unsuccessful at low
shock following cardiac surgery (222– lizing cell membranes and increasing the temperatures, and may actually trigger a
226). Two additional studies have re- likelihood of successful defibrillation if conversion from atrial fibrillation to ven-
ported successful use of hypothermia in arrhythmias do occur. Two studies have tricular fibrillation. Cardioversion for
patients who remained comatose after a assessed the likelihood of successful defi- nonlife-threatening arrhythmias should
massive cardiac arrest leading to severe brillation under hypothermic conditions be avoided at core temperatures ⬍34°C.
cardiac shock (227, 228). Rates of favor- compared with normothermia in a swine Coronary Perfusion and Ischemia.
able outcome in patients with cardiac model. Both found higher rates of return Hypothermia reduces metabolic rate and
shock were 61% (14 of 23) in the first of spontaneous circulation, increased induces bradycardia, thereby providing
study and 39% (11 of 28) in the second likelihood of successful defibrillation, protective effects for the ischemic myo-
study. Oddo et al (229) recently reported fewer shocks required to reach return of cardium. In addition, it has been shown
that the presence of cardiac shock on spontaneous circulation, and lower inci- that mild hypothermia (35°C) induces
admission did not correlate with adverse dence of postdefibrillation asystole dur- coronary vasodilation and increases myo-
outcome in a prospective series of 74 ing mild hypothermia (230, 231). Similar cardial perfusion in healthy volunteers
patients with witnessed cardiac arrest observations have been reported in a rab- (236, 237). In contrast, hypothermia can
treated with hypothermia, regardless of bit model (232). Various case studies de- induce vasoconstriction in severely ath-
initial rhythm or hemodynamic status. scribe successful use of hypothermia to erosclerotic coronary arteries (237). In
These observations suggest that cardiac treat arrhythmias (junctional ectopic addition, hypothermia can cause shiver-

Crit Care Med 2009 Vol. 37, No. 7 (Suppl.) S195


ing, tachycardia, and increased myocar- trolled before cooling is initiated. If this Drugs with long half-lives (e.g., amio-
dial oxygen consumption in insufficiently is not possible, the risks of bleeding darone) are far less affected by these
sedated patients. Indeed, accidental hypo- should be weighed against the benefits of mechanisms, because the amounts elim-
thermia in the perioperative setting in- cooling, and careful consideration should inated in 24 hrs are low regardless of
creases the risk for morbid cardiac events be given to the appropriate depth of hy- temperature; therefore, effects of amioda-
(194, 195). Thus, preventing adverse ef- pothermia for that patient. Very mild hy- rone are less temperature-dependent, and
fects depends on avoiding a stress re- pothermia (35°C) does not affect coagu- clearance is only slightly decreased under
sponse and effective suppression of shiv- lation, and can be safely used even if hypothermic conditions.
ering. bleeding risks are high. Temperatures of Risk for Infections. Hypothermia in-
Some animal experiments and prelim- 33°C to 35°C affect platelet function only; hibits the proinflammatory response
inary clinical studies have suggested that if surgical procedures are performed un- through inhibition of leukocyte migra-
inducing mild hypothermia in the early der hypothermic conditions, platelet tion and phagocytosis and decreased syn-
stages following myocardial infarction transfusion may be considered. Coagula- thesis of proinflammatory cytokines (1).
could mitigate myocardial injury (17). tion factors other than platelet function Indeed, suppression of harmful neuroin-
This issue needs to be addressed in larger are affected only when temperatures de- flammation is one of the mechanisms
studies, but the available evidence cer- crease below 33°C. through which hypothermia may exert its
tainly does not suggest that hypothermia Drug Clearance. The speed of most protective effects; the downside of this
increases myocardial injury. enzyme-mediated reactions is tempera- protective mechanism is an increased
Coagulation. Mild hypothermia can ture-dependent; therefore, the speed of risk for infections. Development of (acci-
induce mild coagulopathy (1). Tempera- these reactions is significantly slowed by dental) hypothermia in the perioperative
tures below 35°C can cause platelet dys- hypothermia. One of the consequences is setting has been linked to increased risk
function and a mild decrease in platelet a decrease in the rates of drug metabo- for infections of the respiratory tract and
count; at temperatures ⬍33°C, other lism by the liver (17, 239). Hypothermia- (surgical) wounds (1, 240 –242). Some ev-
steps in the coagulation cascade, such as induced reductions in clearance have idence suggests that this may even apply
the synthesis and kinetics of clotting en- been demonstrated for a number of com- to controlled normothermia; i.e., sup-
zymes and plasminogen activator inhibi- monly used ICU medications, including pression of a febrile response. For thera-
tors, may also be affected (1). These ef- vasoactive drugs such as epinephrine and peutic cooling (i.e., cooling of sedated
fects have been studied mostly in vitro. norepinephrine; opiates such as mor- and mechanically ventilated patients) the
Clinical observations suggest that the phine, fentanyl, and remifentanil; seda- risk for nosocomial infections appears to
risk for severe bleeding associated with tives such as propofol, volatile anesthet- be closely linked to treatment duration
therapeutic cooling is relatively small; ics, barbiturates, and midazolam; (higher risk if treatment is continued for
none of the studies in patients with car- neuromuscular blocking agents such as ⬎24 hrs) and to the category of patients
diac arrest, severe TBI, or stroke have rocuronium, atracurium, and vecuro- (lowest risk in cardiac arrest patients,
reported significant bleeding problems, nium; and other drugs such as phenytoin, highest risk in patients with severe
although it should be emphasized that nitrates, and some beta-blockers (1, 239). stroke). Most of the studies reporting in-
actively bleeding patients were excluded Other drugs metabolized by the liver are creased risks of infection during thera-
from all of these studies (1, 17). Schefold likely to be affected in similar ways. peutic cooling also observed that final
and coworkers (238) recently assessed the The net effects of hypothermia on outcomes did not appear to be adversely
risk and severity of bleeding during si- drug action may be more complex than affected, even when infections occurred
multaneous use of mild hypothermia and simply increasing the concentration of (17). The suppression of inflammatory re-
thrombolysis. They found that bleeding active metabolites. Temperature can also sponses in the lung has also been used
risks were similar to historical controls directly affect the body’s response to spe- therapeutically to improve oxygenation
treated with thrombolytics alone, al- cific drugs. For example, the effects of in patients with severe acute respiratory
though there was a trend toward more vasoactive drugs, such as epinephrine distress syndrome (17).
red blood cell units being required to and norepinephrine, on blood pressure Strategies to deal with increased infec-
reach target hematocrit in hypothermic can be slightly blunted by hypothermia. tion risks include considering antibiotic
patients who developed bleeding compli- Hypothermia-induced changes in the vol- prophylaxis. Recent publications suggest
cations and needed transfusions. How- ume of distribution and renal function that the use of selective decontamination
ever, neurological outcomes were signif- may also influence how drugs work under of the digestive tract can significantly re-
icantly better in patients treated with hypothermic conditions. duce the risk for nosocomial infections
both thrombolytics and hypothermia, However, in most cases the effect of and decrease mortality in ICU patients
even in those who developed bleeding hypothermia will be to increase drug levels, (243, 244). Clinical studies with thera-
complications (238). The authors con- thereby enhancing drug potency and effect peutic cooling in settings in which selec-
clude that bleeding risks should not be duration. From a practical perspective, it tive decontamination of the digestive
viewed as a reason to withhold hypother- seems reasonable to make empirical dose tract was used have reported low infec-
mia treatment. adjustments in drugs with relatively short tion rates (186, 216), even when hypo-
These factors should be taken into ac- half-lives that are metabolized by the liver thermia was used for prolonged periods
count when deciding whether or not to (e.g., midazolam). When an increase in (216).
use hypothermia in patients who are ac- depth of sedation is required, or an episode Patients should be carefully moni-
tively bleeding, or who have a high bleed- of shivering needs to be treated, it is pref- tored for signs of infection during thera-
ing risk. If possible the (potential) source erable to use bolus doses rather than to peutic temperature management. Some
of bleeding should be (surgically) con- increase maintenance dose. normal signs of infection are absent or

S196 Crit Care Med 2009 Vol. 37, No. 7 (Suppl.)


suppressed during hypothermia; this ob- vic transaminase) and increased levels of in the management of intracranial aneu-
viously applies to development of fever cortisol, norepinephrine, and epineph- rysms. J Neurosurg 1956; 13:1– 42
but may also apply to increased C-reac- rine (184). 6. Botterell EH, Lougheed WM, Morley TP, et
tive protein level and elevated white al: Hypothermia in the surgical treatment
of ruptured intracranial aneurysms. J Neu-
blood cell count. One telltale sign of de- CONCLUSION rosurg 1958; 15:4 –18
veloping infection can be a sudden in- 7. Bigelow WC: Methods for inducing hypo-
crease in the “workload” of the cooling Hypothermia is the first treatment thermia and rewarming. Ann NY Acad Sci
device; i.e., a decrease in water tempera- with a proven ability to reverse postisch- 1959; 80:522–532
ture and increase in noise levels of the emic injury in the clinical setting. As 8. Benson DW, Williams GR, Spencer FC, et al:
device that is being used to cool the pa- such, it represents a major breakthrough, The use of hypothermia after cardiac arrest.
tient. Such increases in cooling power the importance of which is only just be- Anesth Analg 1959; 38:423– 428
may indicate either shivering or an at- ginning to be fully appreciated. However, 9. Williams GR, Spencer FC: The clinical use
tempt by the body to generate fever, and there are numerous pitfalls in using ther- of hypothermia after cardiac arrest. Ann
thus the presence of an infection. apeutic cooling to maximum efficacy, Surg 1959; 148:462– 468
particularly if prolonged treatments are 10. Rosomoff HL: Protective effects of hypo-
Many treatment protocols (including
thermia against pathologic processes of the
in our ICU) include daily blood surveil- required to improve outcome, as seems
nervous system. Ann NY Acad Sci 1959;
lance cultures to screen for bacteremia. to be the case for stroke and severe TBI. 80:475– 486
The threshold for antibiotic treatment Understanding the mechanisms that un- 11. Fay T: Early experiences with local and gen-
should be low. In patients developing in- derlie hypothermia’s protective effects, eral refrigeration of the human brain.
fections after hypothermia treatment, fe- awareness of the physiology of cooling, J Neurosurg 1959; 16:239 –259
ver should be treated symptomatically, to and knowing and understanding the po- 12. Lazorthes G, Campan L: Moderate hypo-
prevent new or additional neurological tential side effects are all key factors in thermia in the treatment of head injuries.
injuries (17). effectively using this therapeutic strat- Clin Neurosurg 1964; 12:293–299
The risk for wound infections may be egy. There are still many misconceptions 13. Hamby WB: Intracranial surgery for aneu-
surrounding therapeutic cooling; some rysm: Effect of hypothermia upon survival.
further increased by hypothermia-in-
J Neurosurg 1963; 20:41– 45
duced cutaneous vasoconstriction. The risks are imagined or overestimated,
14. Safar P: Long-term resuscitation in inten-
same applies to the development of bed- whereas others are not fully appreciated.
sive care units. Anesthesiology 1964; 25:
sores, which once formed will be more Temperature management strategies 216 –222
likely to progress and become infected are likely to play an increasingly impor- 15. Rosomoff HL, Safar P: Management of the
because of the immune-suppressed state tant role in the care of (neuro)critically ill comatose patient. Clin Anesth 1965;
and complete immobilization of the pa- patients. In view of the numerous poten- 1:244 –258
tient. Nurses and physicians should be tial applications for induced hypothermia 16. Bernard SA, Jones BM, Horne MK: Clinical
aware of these additional risks and take or controlled normothermia, it is in- trial of induced hypothermia in comatose
extra precautionary measures. Extra at- creasingly important that physicians and survivors of out-of-hospital cardiac arrest.
tention should also be paid to catheter nurses caring for critically ill patients, Ann Emerg Med 1997; 30:146 –153
particularly those with neurological inju- 17. Polderman KH: Induced hypothermia and
and central line insertion sites, as local
fever control for prevention and treatment
infections also are more likely to develop ries, be aware of these issues, to help
of neurological injuries. Lancet 2008; 371:
here. them avoid the pitfalls and risks of this 1955–1969
Other Effects. Hypothermia is associ- treatment. 18. Jastremski M, Sutton-Tyrrell K, Vaagenes P,
ated with impaired bowel function and et al: Glucocorticoid treatment does not
delayed gastric emptying. There is no REFERENCES improve neurological recovery following
specific strategy to deal with this issue; cardiac arrest. Brain Resuscitation Clinical
routine measures such as insertion of du- 1. Polderman KH, Herold I: Therapeutic hypo- Trial I Study Group. JAMA 1989; 262:
odenal probes and/or administration of thermia and controlled normothermia in 3427–3430
metoclopramide or low-dose erythromy- the intensive care unit: Practical consider- 19. Brain Resuscitation Clinical Trial II Study
ations, side effects, and cooling methods. Group: Randomized clinical study of thio-
cin may be considered, taking care that
Crit Care Med 2009; 37:1101–1120 pental loading in comatose survivors of car-
the combination of hypothermia with diac arrest. N Engl J Med 1986; 314:
2. Fay T: Observations on prolonged human
these drugs does not cause QT prolonga- refrigeration. NY State J Med 1940; 40: 397– 403
tion (1). Hypothermia often causes a mild 1351–1354 20. Brain Resuscitation Clinical Trial II Study
rise in serum amylase; however, the risk 3. Fay T: Observations on generalized refrig- Group: A randomized clinical study of a
for clinically significant pancreatitis is ex- eration in cases of severe cerebral trauma. calcium-entry blocker (lidoflazine) in the
tremely low (1). Assoc Res Nerv Ment Dis Proc 1945; 24: treatment of comatose survivors of cardiac
Many other changes in laboratory 611– 619 arrest. N Engl J Med 1991; 324:1225–1231
measurements can develop during hypo- 4. Bigelow WG, Callaghan JC, Hopps JA: Gen- 21. Helfaer MA, Ichord RN, Martin LJ, et al:
thermia. Apart from those already men- eral hypothermia for experimental intracar- Treatment with the competitive NMDA an-
diac surgery. The use of electrophrenic res- tagonist GPI 3000 does not improve out-
tioned (hyperglycemia; low electrolyte
pirations, an artificial pacemaker for come after cardiac arrest in dogs. Stroke
levels; decreased platelet and white blood
cardiac standstill and radio-frequency re- 1998; 29:824 – 829
cell counts; increased levels of glycerol, warming in general hypothermia. Ann Surg 22. Silbergleit R, Haywood Y, Fiskum G, et al:
free fatty acids, ketonic acids, lactate, and 1950; 132:531–539 Lack of a neuroprotective effect from N-ace-
amylase), these may include a rise in liver 5. Botterell EH, Lougheed WM, Scott JW, et tylcysteine after cardiac arrest and resusci-
enzymes (serum glutamic oxaloacetic al: Hypothermia and interruption of ca- tation in a canine model. Resuscitation
transaminase and serum glutamic pyru- rotid, or carotid and vertebral circulation, 1999; 40:181–186

Crit Care Med 2009 Vol. 37, No. 7 (Suppl.) S197


23. Auer RN: Non-pharmacologic (physiologic) ropoietin on progenitor cell mobilization. induced release of neurotransmitters and
neuroprotection in the treatment of brain J Physiol 2008; 586(Pt 23):5757–5769 free fatty acids in rat brain. Stroke 1989;
ischemia. Ann NY Acad Sci 2001; 939: 39. Povlishock JT, Buki A, Koiziumi H, et al: 20:904 –910
271–282 Initiating mechanisms involved in the 53. Kaibara T, Sutherland GR, Colbourne F, et
24. Corbett D, Nurse S: The problem of assess- pathobiology of traumatically induced ax- al: Hypothermia: Depression of tricarboxy-
ing effective neuroprotection in experimen- onal injury and interventions targeted at lic acid cycle flux and evidence for pentose
tal cerebral ischemia. Prog Neurobiol 1998; blunting their progression. Acta Neurochir phosphate shunt upregulation. J Neurosurg
54:531–548 Suppl (Wien) 1999; 73:15–20 1999; 90:339 –347
25. Siesjo BK, Bengtsson F, Grampp W, et al: 40. Xu L, Yenari MA, Steinberg GK, et al: Mild 54. Kuboyama K, Safar P, Radovsky A, et al:
Calcium, excitotoxins, and neuronal death hypothermia reduces apoptosis of mouse Delay in cooling negates the beneficial ef-
in brain. Ann NY Acad Sci 1989; 568: neurons in vitro early in the cascade. fect of mild resuscitative cerebral hypother-
234 –251 J Cereb Blood Flow Metab 2002; 22:21–28 mia after cardiac arrest in dogs: A prospec-
26. Dietrich WD, Busto R, Alonso O, et al: Intra- 41. Adachi M, Sohma O, Tsuneishi S, et al: tive, randomized study. Crit Care Med 1993;
ischemic but not post-ischemic brain hypo- Combination effect of systemic hypother- 21:1348 –1358
thermia protects chronically following mia and caspase inhibitor administration 55. Baker CJ, Onesti ST, Solomon RA: Reduc-
global forebrain ischemia in rats. J Cereb against hypoxic-ischemic brain damage in tion by delayed hypothermia of cerebral in-
Blood Flow Metab 1993; 13:541–549 neonatal rats. Pediatr Res 2001; 50: farction following middle cerebral artery
27. Small DL, Morley P, Buchan AM: Biology of 590 –595 occlusion in the rat: A time-course study.
ischemic cerebral cell death. Prog Cardio- 42. Ning XH, Chen SH, Xu CS, et al: Hypother- J Neurosurg 1992; 77:438 – 444
vasc Dis 1999; 42:185–207 mic protection of the ischemic heart via 56. Hickey RW, Ferimer H, Alexander HL, et al:
28. Polderman KH: Application of therapeutic alterations in apoptotic pathways as as- Delayed, spontaneous hypothermia reduces
hypothermia in the ICU: Opportunities and sessed by gene array analysis. J Appl Physiol neuronal damage after asphyxial cardiac ar-
pitfalls of a promising treatment modality. 2002; 92:2200 –2207 rest in rats. Crit Care Med 2000; 28:
Part 1: Indications and evidence. Intensive 43. Liou AK, Clark RS, Henshall DC, et al: To 3511–3516
Care Med 2004; 30:556 –575 die or not to die for neurons in ischemia, 57. Coimbra C, Wieloch T: Hypothermia ame-
29. Milde LN: Clinical use of mild hypothermia traumatic brain injury and epilepsy: A re- liorates neuronal survival when induced 2
for brain protection: A dream revisited. view on the stress-activated signaling path- hours after ischaemia in the rat. Acta
J Neurosurg Anesthesiol 1992; 4:211–215 ways and apoptotic pathways. Prog Neuro- Physiol Scand 1992; 146:543–544
30. Takata K, Takeda Y, Morita K: Effects of biol 2003; 69:103–142
58. Colbourne F, Li H, Buchan AM: Indefatiga-
hypothermia for a short period on histolog- 44. Raghupathi R, Graham DI, McIntosh TK:
ble CA1 sector neuroprotection with mild
ical outcome and extracellular glutamate Apoptosis after traumatic brain injury.
hypothermia induced 6 hours after severe
concentration during and after cardiac ar- J Neurotrauma 2000; 17:927–938
forebrain ischemia in rats. J Cereb Blood
rest in rats. Crit Care Med 2005; 33: 45. Leker RR, Shohami E: Cerebral ischemia
Flow Metab 1999; 19:742–749
1340 –1345 and trauma— different etiologies yet simi-
59. Coimbra C, Wieloch T: Moderate hypother-
31. Hagerdal M, Harp J, Nilsson L, et al: The lar mechanisms: Neuroprotective opportu-
mia mitigates neuronal damage in the rat
effect of induced hypothermia upon oxygen nities. Brain Res Brain Res Rev 2002; 39:
brain when initiated several hours following
consumption in the rat brain. J Neurochem 55–73
transient cerebral ischemia. Acta Neuro-
1975; 24:311–316 46. Hall ED: Free radicals in stroke. In: Stroke
pathol 1994; 87:325–333
32. Palmer C, Vannucci RC, Christensen MA, et Therapy: Basic, Preclinical, and Clinical Di-
60. Leonov Y, Sterz F, Safar P, et al: Moderate
al: Regional cerebral blood flow and glucose rections. Miller LP (Ed). New York, Wiley-
hypothermia after cardiac arrest of 17 min-
utilization during hypothermia in newborn Liss, 1999, pp 245–270
utes in dogs: Effect on cerebral and cardiac
dogs. Anesthesiology 1989; 71:730 –737 47. Globus MY-T, Busto R, Lin B, et al: Detec-
33. Aoki M, Nomura F, Stromski ME, et al: tion of free radical activity during transient outcome: A preliminary study. Stroke 1990;
Effects of pH on brain energetics after hy- global ischemia and recirculation: Effects of 21:1600 –1606
pothermic circulatory arrest. Ann Thorac intra-ischemic brain temperature modula- 61. Leonov Y, Sterz F, Safar P, et al: Mild cere-
Surg 1993; 55:1093–1103 tion. J Neurochem 1995; 65:1250 –1256 bral hypothermia during and after cardiac
34. Ehrlich MP, McCullough JN, Zhang N, et al: 48. Dempsey RJ, Combs DJ, Maley ME, et al: arrest improves neurologic outcome in
Effect of hypothermia on cerebral blood Moderate hypothermia reduces postisch- dogs. J Cereb Blood Flow Metab 1990; 10:
flow and metabolism in the pig. Ann Thorac emic edema development and leukotriene 57–70
Surg 2002; 73:191–197 production. Neurosurgery 1987; 21: 62. Sterz F, Safar P, Tisherman S, et al: Mild
35. Erecinska M, Thoresen M, Silver IA: Effects 177–181 hypothermic cardiopulmonary resuscita-
of hypothermia on energy metabolism in 49. Busto R, Dietrich WD, Globus MY, et al: tion improves outcome after prolonged car-
mammalian central nervous system. Small differences in intraischemic brain diac arrest in dogs. Crit Care Med 1991;
J Cereb Blood Flow Metab 2003; 23: temperature critically determine the extent 19:379 –389
513–530 of ischemic neuronal injury. J Cereb Blood 63. Safar P, Xiao F, Radovsky A, et al: Improved
36. Coles JP, Minhas PS, Fryer TD, et al: Effect Flow Metab 1987; 7:729 –738 cerebral resuscitation from cardiac arrest in
of hyperventilation on cerebral blood flow 50. Globus MY-T, Alonso O, Dietrich WD, et al: dogs with mild hypothermia plus blood flow
in traumatic head injury: Clinical relevance Glutamate release and free radical produc- promotion. Stroke 1996; 27:105–113
and monitoring correlates. Crit Care Med tion following brain injury: Effects of post- 64. Kollmar R, Schabitz WR, Heiland S, et al:
2002; 30:1950 –1959 traumatic hypothermia. J Neurochem 1995; Neuroprotective effect of delayed moderate
37. Coles JP, Fryer TD, Coleman MR, et al: 65:1704 –1711 hypothermia after focal cerebral ischemia:
Hyperventilation following head injury: Ef- 51. Baker AJ, Zornow MH, Grafe MR, et al: An MRI study. Stroke 2002; 33:1899 –1904
fect on ischemic burden and cerebral oxi- Hypothermia prevents ischemia-induced 65. Weinrauch V, Safar P, Tisherman S, et al:
dative metabolism. Crit Care Med 2007; 35: increases in hippocampal glycine concen- Beneficial effect of mild hypothermia and
568 –578 trations in rabbits. Stroke 1991; 22: detrimental effect of deep hypothermia after
38. Lin JS, Chen YS, Chiang HS, et al: Hypoxic 666 – 673 cardiac arrest in dogs. Stroke 1992; 23:
preconditioning protects rat hearts against 52. Busto R, Globus MY, Dietrich WD, et al: 1454 –1462
ischemia/reperfusion injury: Role of eryth- Effect of mild hypothermia on ischemia- 66. Gisvold SE, Safar P, Rao G, et al: Multifac-

S198 Crit Care Med 2009 Vol. 37, No. 7 (Suppl.)


eted therapy after global brain ischemia in pothermia. Can J Neurol Sci 1999; 26: hippocampal neurons. Exp Neurol 2000;
monkeys. Stroke 1984; 15:803– 812 298 –304 162:1–12
67. Markarian GZ, Lee JH, Stein DJ, et al: Mild 82. Chi OZ, Liu X, Weiss HR: Effects of mild 99. Natale JA, D’Alecy LG: Protection from ce-
hypothermia: Therapeutic window after ex- hypothermia on blood– brain barrier dis- rebral ischemia by brain cooling without
perimental cerebral ischemia. Neurosur- ruption during isoflurane or pentobarbital reduced lactate accumulation in dogs.
gery 1996; 38:542–550 anesthesia. Anesthesiology 2001; 95: Stroke 1989; 20:770 –777
68. Dietrich WD, Busto R, Globus MY, et al: 933–938 100. Jiang JY, Liang YM, Luo QZ, et al: Effect of
Brain damage and temperature: Cellular 83. Smith SL, Hall ED: Mild pre- and posttrau- mild hypothermia on brain dialysate lactate
and molecular mechanisms. Adv Neurol matic hypothermia attenuates blood– brain after fluid percussion brain injury in ro-
1996; 71:177–194 barrier damage following controlled corti- dents. Neurosurgery 2004; 54:713–717
69. Schmidt OI, Heyde CE, Ertel W, et al: cal impact injury in the rat. J Neurotrauma 101. Chatauret N, Rose C, Therrien G, et al: Mild
Closed head injury—an inflammatory dis- 1996; 13:1–9 hypothermia prevents cerebral edema and
ease? Brain Res Brain Res Rev 2005; 48: 84. Jurkovich GJ, Pitt RM, Curreri PW, et al: CSF lactate accumulation in acute liver fail-
388 –399 Hypothermia prevents increased capillary ure. Metab Brain Dis 2001; 16:95–102
70. Asensio VC, Campbell IL: Chemokines in permeability following ischemia–reperfusion 102. Amess PN, Penrice J, Cady EB, et al: Mild
the CNS: Plurifunctional mediators in di- injury. J Surg Res 1988; 44:514 –521 hypothermia after severe transient hypoxia—
verse states. Trends Neurosci 1999; 22: 85. Kinoshita K, Chatzipanteli K, Alonso OF, et ischemia reduces the delayed rise in cere-
504 –512 al: The effect of brain temperature on he- bral lactate in the newborn piglet. Pediatr
71. Merrill JE, Benveniste EN: Cytokines in in- moglobin extravasation after traumatic Res 1997; 41:803– 808
flammatory brain lesions: Helpful and brain injury. J Neurosurg 2002; 97:945–953 103. Vejchapipat P, Proctor E, Ramsay A, et al:
harmful. Trends Neurosci 1996; 19:331–338 86. Fischer S, Clauss M, Wiesnet M, et al: Hyp- Intestinal energy metabolism after isch-
72. Morganti-Kossmann MC, Rancan M, Stahel oxia induces permeability in brain mi- emia-reperfusion: Effects of moderate hypo-
PF, et al: Inflammatory response in acute crovessel endothelial cells via VEGF and thermia and perfluorocarbons. J Pediatr
traumatic brain injury: A double-edged NO. Am J Physiol 1999; 276:C812–C820 Surg 2002; 37:786 –790
sword. Curr Opin Crit Care 2002; 87. Fischer S, Renz D, Wiesnet M, et al: Hypo- 104. Chatauret N, Zwingmann C, Rose C, et al:
8:101–105 thermia abolishes hypoxia-induced hyper- Effects of hypothermia on brain glucose
73. Schoettle RJ, Kochanek PM, Magargee MJ, permeability in brain microvessel endothe- metabolism in acute liver failure: A H/C-
et al: Early polymorphonuclear leukocyte lial cells. Brain Res Mol Brain Res 1999; nuclear magnetic resonance study. Gastro-
74:135–144
accumulation correlates with the develop- enterology 2003; 125:815– 824
88. Polderman KH, Ely EW, Badr AE, et al:
ment of posttraumatic cerebral edema in 105. Vaquero J, Blei AT: Mild hypothermia for
Induced hypothermia in traumatic brain in-
rats. J Neurotrauma 1990; 7:207–217 acute liver failure: A review of mechanisms
jury: Considering the conflicting results of
74. Patel HC, Boutin H, Allan SM: Interleukin-1 of action. J Clin Gastroenterol 2005; 39:
meta-analyses and moving forward. Inten-
in the brain: Mechanisms of action in acute S147–S157
sive Care Med 2004; 30:1860 –1864
neurodegeneration. Ann NY Acad Sci 2003; 106. Lanier WL: Cerebral metabolic rate and hy-
89. Ghajar J: Traumatic brain injury. Lancet
992:39 – 47 pothermia: Their relationship with isch-
2000; 356:923–929
75. Aibiki M, Maekawa S, Ogura S, et al: Effect emic neurologic injury. J Neurosurg Anes-
90. Reinert MM, Bullock R: Clinical trials in
of moderate hypothermia on systemic and thesiol 1995; 7:216 –221
head injury. Neurol Res 1999; 21:330 –338
internal jugular plasma IL-6 levels after 107. Soukup J, Zauner A, Doppenberg EM, et al:
91. Yokota S, Imagawa T, Miyamae T, et al:
traumatic brain injury in humans. J Neuro- Relationship between brain temperature,
Hypothetical pathophysiology of acute en-
trauma 1999; 16:225–232 brain chemistry and oxygen delivery after
cephalopathy and encephalitis related to in-
76. Kimura A, Sakurada S, Ohkuni H, et al: severe human head injury: The effect of
fluenza virus infection and hypothermia
Moderate hypothermia delays proinflamma- therapy. Pediatr Int 2000; 42:197–203 mild hypothermia. Neurol Res 2002; 24:
tory cytokine production of human periph- 92. Morimoto Y, Kemmotsu O, Kitami K, et al: 161–168
eral blood mononuclear cells. Crit Care Acute brain swelling after out-of-hospital 108. Kimura T, Sako K, Tanaka K, et al: Effect of
Med 2002; 30:1499 –1502 cardiac arrest: Pathogenesis and outcome. mild hypothermia on energy state recovery
77. Suehiro E, Fujisawa H, Akimura T, et al: In- Crit Care Med 1993; 21:104 –110 following transient forebrain ischemia in
creased matrix metalloproteinase-9 in blood 93. Brain Trauma Foundation: Guidelines for the gerbil. Exp Brain Res 2002; 145:83–90
in association with activation of interleukin-6 the management of severe traumatic brain 109. Nybo L, Secher NH, Nielsen B: Inadequate
after traumatic brain injury: Influence of hy- injury. http://www.braintrauma.org. Ac- heat release from the human brain during
pothermic therapy. J Neurotrauma 2004; 21: cessed May 13, 2009. prolonged exercise with hyperthermia.
1706 –1711 94. Stocchetti N, Colombo A, Ortolano F, et al: J Physiol 2002; 545:697–704
78. Dietrich WD, Chatzipanteli K, Vitarbo E, et Time course of intracranial hypertension 110. Mariak Z, Bondyra Z, Piekarska M: The tem-
al: The role of inflammatory processes in after traumatic brain injury. J Neuro- perature within the circle of Willis versus
the pathophysiology and treatment of brain trauma 2007; 24:1339 –1346 tympanic temperature in resting normo-
and spinal cord trauma. Acta Neurochir 95. Clifton GL, Miller ER, Choi SC, et al: Lack thermic humans. Eur J Appl Physiol Occup
Suppl 2004; 89:69 –74 of effect of induction of hypothermia after Physiol 1993; 66:518 –520
79. Novack TA, Dillon MC, Jackson WT: Neuro- acute brain injury. N Engl J Med 2001; 111. Crowder CM, Tempelhoff R, Theard MA, et
chemical mechanisms in brain injury and 344:556 –563 al: Jugular bulb temperature: Comparison
treatment: A review. J Clin Exp Neuropsy- 96. McIntyre LA, Fergusson DA, Hebert PC, et with brain surface and core temperatures in
chol 1996; 18:685–706 al: Prolonged therapeutic hypothermia after neurosurgical patients during mild hypo-
80. Raghupathi R, McIntosh TK: Pharmaco- traumatic brain injury in adults: A system- thermia. J Neurosurg 1996; 85:98 –103
therapy for traumatic brain injury: A re- atic review. JAMA 2003; 289:2992–2999 112. Mcilvoy L: Comparison of brain tempera-
view. Proc West Pharmacol Soc 1998; 41: 97. Schaller B, Graf R: Hypothermia and stroke: ture to core temperature: A review of the
241–246 The pathophysiological background. Patho- literature. J Neurosci Nurs 2004; 36:23–31
81. Huang ZG, Xue D, Preston E, et al: Biphasic physiology 2003; 10:7–35 113. Henker RA, Brown SD, Marion DW: Com-
opening of the blood– brain barrier follow- 98. Ding D, Moskowitz SI, Li R, et al: Acidosis parison of brain temperature with bladder
ing transient focal ischemia: Effects of hy- induces necrosis and apoptosis of cultured and rectal temperatures in adults with se-

Crit Care Med 2009 Vol. 37, No. 7 (Suppl.) S199


vere head injury. Neurosurgery 1998; 42: 130. Wass CT, Lanier WL, Hofer RE, et al: Tem- and the role of coagulation activation after
1071–1075 perature changes of ⬎ or ⫽ 1°C alter func- cardiac arrest. Curr Opin Crit Care 2001;
114. Rumana CS, Gopinath SP, Uzura M, et al: tional neurologic outcome and histopathol- 7:176 –183
Brain temperature exceeds systemic tem- ogy in a canine model of complete cerebral 145. Gando S, Kameue T, Nanzaki S, et al: Mas-
perature in head-injured patients. Crit Care ischemia. Anesthesiology 1995; 83:325–335 sive fibrin formation with consecutive im-
Med 1998; 26:562–567 131. Baena RC, Busto R, Dietrich WD, et al: pairment of fibrinolysis in patients with
115. Verlooy J, Heytens L, Veeckmans G, et al: Hyperthermia delayed by 24 hours aggra- out-of-hospital cardiac arrest. Thromb Hae-
Intracerebral temperature monitoring in vates neuronal damage in rat hippocampus most 1997; 77:278 –282
severely head injured patients. Acta Neuro- following global ischemia. Neurology 1997; 146. Fischer M, Bottiger BW, Popov-Cenic S, et
chir (Wien) 1995; 134:76 –78 48:768 –773 al: Thrombolysis using plasminogen activa-
116. Mellergard P, Nordstrom CH: Intracerebral 132. Kim Y, Busto R, Dietrich WD, et al: Delayed tor and heparin reduces cerebral no-reflow
temperature in neurosurgical patients. postischemic hyperthermia in awake rats after resuscitation from cardiac arrest: An
Neurosurgery 1991; 28:709 –713 worsens the histopathological outcome of experimental study in the cat. Intensive
117. Schwab S, Spranger M, Aschhoff A, et al: transient focal cerebral ischemia. Stroke Care Med 1996; 22:1214 –1223
Brain temperature monitoring and modu- 1996; 27:2274 –2281 147. Darius H, Yanagisawa A, Brezinski ME, et
lation in patients with severe MCA infarc- 133. Hickey RW, Kochanek PM, Ferimer H, et al: al: Beneficial effects of tissue-type plasmin-
tion. Neurology 1997; 48:762–767 Induced hyperthermia exacerbates neuro- ogen activator in acute myocardial ischemia
118. Childs C, Vail A, Protheroe R, et al: Differ- logic neuronal histologic damage after as- in cats. J Am Coll Cardiol 1986; 8:125–131
ences between brain and rectal tempera- phyxial cardiac arrest in rats. Crit Care Med 148. Kim YH, Park JH, Hong SH, et al: Nonpro-
tures during routine critical care of patients 2003; 31:531–535 teolytic neuroprotection by human recom-
with severe traumatic brain injury. Anaes- 134. Chen H, Chopp M, Welch KMA: Effect of binant tissue plasminogen activator. Sci-
thesia 2005; 60:759 –765 mild hyperthermia on the ischemic infarct ence 1999; 284:647– 650
119. Albrecht RF, Wass CT, Lanier WL: Occur- volume after middle cerebral artery occlu- 149. Böttiger BW, Bode C, Kern S, et al: Efficacy
rence of potentially detrimental tempera- sion in the rat. Neurology 1991; 41: and safety of thrombolytic therapy after ini-
ture alterations in hospitalized patients at 1133–1135 tially unsuccessful cardiopulmonary resus-
risk for brain injury. Mayo Clin Proc 1998; 135. Wass CT, Lanier WL, Hofer RE, et al: Tem- citation: A prospective clinical trial. Lancet
73:629 – 635 perature changes of ⬎ or ⫽ 1 degree C alter 2001; 357:1583–1585
120. Albrecht RF 2nd, Wass CT, Lanier WL, et al: functional neurologic outcome and histopa- 150. Spöhr F, Arntz HR, Bluhmki E, et al: Inter-
Uncontrolled hyperthermia in the neuro- thology in a canine model of complete ce-
national multicentre trial protocol to assess
surgical intensive care unit. Neurosurgery rebral ischemia. Anesthesiology 1995; 83:
the efficacy and safety of tenecteplase dur-
2000; 47:850 – 855 325—335
ing cardiopulmonary resuscitation in pa-
121. Schwarz S, Hafner K, Aschoff A, et al: Inci- 136. Ginsberg MD, Sternau LL, Globus MY-T, et
tients with out-of-hospital cardiac arrest:
dence and prognostic significance of fever al: Therapeutic modulation of brain tem-
The Thrombolysis in Cardiac Arrest
following intracerebral hemorrhage. Neu- perature: Relevance to ischemic brain in-
(TROICA) Study. Eur J Clin Invest 2005;
rology 2000; 54:354 –361 jury. Cerebrovasc Brain Metab Rev 1992;
35:315–323
122. Reith J, Jørgensen HS, Pedersen PM, et al: 4:189 –225
151. Valeri CR, Feingold H, Cassidy G, et al:
Body temperature in acute stroke: Relations 137. Takino M, Okada Y: Hyperthermia following
Hypothermia-induced reversible platelet
to stroke severity, infarct size, mortality, cardiopulmonary resuscitation. Intensive
dysfunction. Ann Surg 1987; 205:175–181
and outcome. Lancet 1996; 347:422– 425 Care Med 1991; 17:419 – 420
152. Michelson AD, MacGregor H, Barnard MR,
123. Hajat C, Hajat S, Sharma P: Effects of post- 138. Zeiner A, Holzer M, Sterz F, et al: Hyper-
et al: Hypothermia-induced reversible
stroke pyrexia on stroke outcome: Meta- thermia after cardiac arrest is associated
platelet dysfunction. Thromb Haemost
analysis of studies on patients. Stroke 2000; with an unfavorable neurologic outcome.
31:410 – 414 Arch Intern Med 2001; 161:2007–2012 1994; 71:633– 640
124. Gorbach AM, Heiss J, Kufta C, et al: Intra- 139. Soukup J, Zauner A, Doppenberg EM, et al: 153. Valeri CR, MacGregor H, Cassidy G, et al:
operative infrared functional imaging of hu- The importance of brain temperature in pa- Effects of temperature on bleeding time and
man brain. Ann Neurol 2003; 54:297–309 tients after severe head injury: Relationship clotting time in normal male and female
125. Collins CM, Smith MB, Turner R: Model of to intracranial pressure, cerebral perfusion volunteers. Crit Care Med 1995; 23:
local temperature changes in brain upon pressure, cerebral blood flow, and outcome. 698 –704
functional activation. J Appl Physiol 2004; J Neurotrauma 2002; 19:559 –571 154. Watts DD, Trask A, Soeken K, et al: Hypo-
97:2051–2055 140. Azzimondi G, Bassein L, Nonino F, et al: thermic coagulopathy in trauma: Effect of
126. Gorbach AM: Infrared imaging of brain Fever in acute stroke worsens prognosis. A varying levels of hypothermia on enzyme
function. Adv Exp Med Biol 1993; 333: prospective study. Stroke 1995; 26: speed, platelet function, and fibrinolytic ac-
95–123 2040 –2043 tivity. J Trauma 1998; 44:846 – 854
127. Ecker RD, Goerss SJ, Meyer FB, et al: Vision 141. Castillo J, Davalos A, Marrugat J, et al: Timing 155. Leffer CW: Prostanoids: Intrinsic modula-
of the future: Initial experience with intra- for fever-related brain damage in acute isch- tion of cerebral circulation. News Physiol
operative real-time high-resolution dy- emic stroke. Stroke 1998; 29:2455–2460 Sci 1997; 12:72–77
namic infrared imaging. Technical note. 142. Kammersgaard LP, Jorgensen HS, Rungby 156. Dogne JM, de Leval X, Hanson J, et al: New
J Neurosurg 2002; 97:1460 –1471 JA, et al: Admission body temperature pre- developments on thromboxane and prosta-
128. Hayashi N, Hirayama T, Udagawa A, et al: dicts long-term mortality after acute stroke: cyclin modulators part I: Thromboxane
Systemic management of cerebral edema The Copenhagen Stroke Study. Stroke modulators. Curr Med Chem 2004; 11:
based on a new concept in severe head in- 2002; 33:1759 –1762 1223–1241
jury patients. Acta Neurochir Suppl (Wien) 143. Böttiger BW, Motsch J, Bohrer H, et al: 157. De Leval X, Hanson J, David JL, et al: New
1994; 60:541–543 Activation of blood coagulation after cardiac developments on thromboxane and prosta-
129. Piepgras A, Elste V, Frietsch T, et al: Effect of arrest is not balanced adequately by activa- cyclin modulators part II: Prostacyclin
moderate hypothermia on experimental se- tion of endogenous fibrinolysis. Circulation modulators. Curr Med Chem 2004; 11:
vere subarachnoid hemorrhage, as evaluated 1995; 92:2572–2578 1243–1252
by apparent diffusion coefficient changes. 144. Böttiger BW, Martin E: Thrombolytic ther- 158. Westcott JY, Murphy RC, Stenmark K: Ei-
Neurosurgery 2001; 48:1128 –1135 apy during cardiopulmonary resuscitation cosanoids in human ventricular cerebrospi-

S200 Crit Care Med 2009 Vol. 37, No. 7 (Suppl.)


nal fluid following severe brain injury. Pros- seizures: An electroencephalographic and mic arrest influence the outcome in a swine
taglandins 1987; 34:877– 887 14C-deoxyglucose autoradiographic study. model of lethal hemorrhage? J Trauma
159. Hsu CY, Halushka PV, Hogan EL, et al: Brain Res 1999; 818:228 –235 2006; 60:134 –146
Alteration of thromboxane and prostacyclin 174. Karkar KM, Garcia PA, Bateman LM, et al: 189. Hildebrand F, van Griensven M, Giannoudis
levels in experimental spinal cord injury. Focal cooling suppresses spontaneous epi- P, et al: Effects of hypothermia and re-
Neurology 1985; 35:1003–1009 leptiform activity without changing the cor- warming on the inflammatory response in a
160. Chen ST, Hsu CY, Hogan EL, et al: Throm- tical motor threshold. Epilepsia 2002; 43: murine multiple hit model of trauma. Cy-
boxane, prostacyclin, and leukotrienes in 932–935 tokine 2005; 31:382–393
cerebral ischemia. Neurology 1986; 36: 175. Corry JJ, Dhar R, Murphy T, et al: Hypo- 190. Kawahara F, Kadoi Y, Saito S, et al: Slow
466 – 470 thermia for refractory status epilepticus. rewarming improves jugular venous oxygen
161. Maekawa S, Aibiki M, Ogura S, et al: Mild Neurocrit Care 2008; 9:189 –197 saturation during rewarming. Acta Anaes-
hypothermia suppresses thromboxane B2 176. Lundgren J, Smith ML, Blennow G, et al: thesiol Scand 2003; 47:419 – 424
production in brain-injured patients. In: Hyperthermia aggravates and hypothermia 191. Bissonnette B, Holtby HM, Davis AJ, et al:
The Immune Consequences of Trauma, ameliorates epileptic brain damage. Exp Cerebral hyperthermia in children after car-
Shock and Sepsis. Mechanisms and Thera- Brain Res 1994; 99:43–55 diopulmonary bypass. Anesthesiology 2000;
peutic Approaches. Faist E (Ed). Bologna, 177. Yager JY, Armstrong EA, Jaharus C, et al: 93:611– 618
Italy, Monduzzi Editore, 1997, pp 135–138 Preventing hyperthermia decreases brain 192. Lavinio A, Timofeev I, Nortje J, et al: Cere-
162. Aibiki M, Maekawa S, Yokono S: Moderate damage following neonatal hypoxic-isch- brovascular reactivity during hypothermia
hypothermia improves imbalances of emic seizures. Brain Res 2004; 1011:48 –57 and rewarming. Br J Anaesth 2007; 99:
thromboxane A2 and prostaglandin I2 pro- 178. Liu Z, Gatt A, Mikati M, et al: Effect of 237–244
duction after traumatic brain injury in hu- temperature on kainic acid-induced sei- 193. Sessler DI: Thermoregulatory defense
mans. Crit Care Med 2000; 28:3902–3906 zures. Brain Res 1993; 631:51–58 mechanisms. Crit Care Med 2009; 37:
163. Chen L, Piao Y, Zeng F, et al: Moderate 179. Somjen GG, Aitken PG, Balestrino M, et al: S203–S210
hypothermia therapy for patients with se- Spreading depression-like depolarization 194. Frank SM, Fleisher LA, Breslow MJ, et al:
vere head injury. Chin J Traumatol 2001; and selective vulnerability of neurons. A Perioperative maintenance of normother-
4:164 –167 brief review. Stroke 1990; 21(Suppl):III- mia reduces the incidence of morbid car-
164. Yuan HB, Huang Y, Zheng S, et al: Hypo- 179 –III-183 diac events. A randomized clinical trial.
thermic preconditioning increases survival 180. Lundgren J, Smith ML, Siesjo BK: Influ- JAMA 1997; 277:1127–1134
of purkinje neurons in rat cerebellar slices ence of moderate hypothermia on ischemic 195. Frank SM, Beattie C, Christopherson R, et
after an in vitro simulated ischemia. Anes- brain damage incurred under hyperglyce- al: Unintentional hypothermia is associated
thesiology 2004; 100:331–337 mic conditions. Exp Brain Res 1991; 84: with postoperative myocardial ischemia.
165. Yunoki M, Nishio S, Ukita N, et al: Hypo- 91–101 The Perioperative Ischemia Randomized
thermic preconditioning induces rapid tol- 181. Church AJ, Andrew RD: Spreading depres- Anesthesia Trial Study Group. Anesthesiol-
erance to focal ischemic injury in the rat. sion expands traumatic injury in neocorti- ogy 1993; 78:468 – 476
Exp Neurol 2003; 181:291–300 cal brain slices. J Neurotrauma 2005; 22: 196. Leslie K, Sessler DI: Perioperative hypo-
166. Strauch JT, Lauten A, Spielvogel D, et al: 277–290 thermia in the high-risk surgical patient.
Mild hypothermia protects the spinal cord 182. Yenari MA, Onley D, Hedehus M, et al: Dif- Best Pract Res Clin Anaesthesiol 2003; 17:
from ischemic injury in a chronic porcine fusion- and perfusion-weighted magnetic 485– 498
model. Eur J Cardiothorac Surg 2004; 25: resonance imaging of focal cerebral isch- 197. De Witte J, Sessler DI: Perioperative shiver-
708 –715 emia and cortical spreading depression un- ing: Physiology and pharmacology. Anes-
167. Oku K, Kuboyama K, Safar P, et al: Cerebral der conditions of mild hypothermia. Brain thesiology 2002; 96:467– 484
and systemic arteriovenous oxygen moni- Res 2000; 885:208 –219 198. Van Zanten ARH, Polderman KH: Skin
toring after cardiac arrest: Inadequate cere- 183. Polderman KH, Callaghan J: Equipment re- counterwarming to prevent shivering dur-
bral oxygen delivery. Resuscitation 1994; view: Cooling catheters to induce therapeu- ing therapeutic cooling: Blowing hot and
27:141–152 tic hypothermia? Crit Care 2006; 10:234 cold? Crit Care Med 2009; 37:2106 –2108
168. Jordan KG: Nonconvulsive status epilepti- 184. Polderman KH: Application of therapeutic 199. Badjatia N, Strongilis E, Prescutti M, et al:
cus in acute brain injury. J Clin Neuro- hypothermia in the intensive care unit. Op- Metabolic benefits of surface counter warm-
physiol 1999; 16:332–340 portunities and pitfalls of a promising treat- ing during therapeutic temperature modu-
169. Krumholz A: Epidemiology and evidence ment modality. Part 2: Practical aspects and lation. Crit Care Med 2009; 37:1893–1897
for morbidity of nonconvulsive status epi- side effects. Intensive Care Med 2004; 30: 200. Badjatia N, Strongilis E, Gordon E, et al:
lepticus. J Clin Neurophysiol 1999; 16: 757–1769 Metabolic impact of shivering during ther-
314 –322 185. Polderman KH, Peerdeman SM, Girbes ARJ: apeutic temperature modulation. The Bed-
170. Vespa PM, Nuwer MR, Nenov V, et al: In- Hypophosphatemia and hypomagnesemia side Shivering Assessment Scale. Stroke
creased incidence and impact of nonconvul- induced by cooling in patients with severe 2008; 39:3242–3247
sive and convulsive seizures after traumatic head injury. J Neurosurg 2001; 94:697–705 201. Thoresen M, Satas S, Loberg EM, et al:
brain injury as detected by continuous elec- 186. Polderman KH, Rijnsburger ER, Peerdeman Twenty-four hours of mild hypothermia in
troencephalographic monitoring. J Neuro- SM, et al: Induction of hypothermia in pa- unsedated newborn pigs starting after a se-
surg 1999; 91:750 –760 tients with various types of neurologic in- vere global hypoxic–ischemic insult is not
171. Towne AR, Waterhouse EJ, Boggs JG, et al: jury with use of large volumes of ice-cold neuroprotective. Pediatr Res 2001; 50:
Prevalence of nonconvulsive status epilep- intravenous fluid. Crit Care Med 2005; 33: 405– 411
ticus in comatose patients. Neurology 2000; 2744 –2751 202. Tooley JR, Satas S, Porter H, et al: Head
54:340 –345 187. Maxwell WL, Watson A, Queen R, et al: cooling with mild systemic hypothermia in
172. Dennis LJ, Claassen J, Hirsch LJ, et al: Non- Slow, medium, or fast re-warming follow- anesthetized piglets is neuroprotective. Ann
convulsive status epilepticus after sub- ing post-traumatic hypothermia therapy? Neurol 2003; 53:65–72
arachnoid hemorrhage. Neurosurgery An ultrastructural perspective. J Neuro- 203. Bacher A: Effects of body temperature on
2002; 51:1136 –1143 trauma 2005; 22:873– 884 blood gases. Intensive Care Med 2005; 31:
173. Maeda T, Hashizume K, Tanaka T: Effect of 188. Alam HB, Rhee P, Honma K, et al: Does the 24 –27
hypothermia on kainic acid-induced limbic rate of rewarming from profound hypother- 204. Polderman KH, van Zanten ARH, Girbes

Crit Care Med 2009 Vol. 37, No. 7 (Suppl.) S201


ARJ: The importance of magnesium in crit- Mechanism of cold diuresis in the rat. Am J Moderate hypothermia increases the chance
ically ill patients: A role in mitigating neu- Physiol 1983; 244:F210 –F216 of spiral wave collision in favor of self-
rological injury and in the prevention of 219. Allen DE, Gellai M: Mechanisms for the termination of ventricular tachycardia/
vasospasms. Intensive Care Med 2003; 29: diuresis of acute cold exposure: Role for fibrillation. Am J Physiol Heart Circ Physiol
1202–1203 vasopressin?. Am J Physiol 1993; 264: 2008; 294:H1896 –H1905
205. Soliman HM, Mercan D, Lobo SS, et al: R524 –R532 233. Bash SE, Shah JJ, Albers WH, et al: Hypo-
Development of ionized hypomagnesemia is 220. Sun Z: Genetic AVP deficiency abolishes cold- thermia for the treatment of postsurgical
associated with higher mortality rates. Crit induced diuresis but does not attenuate cold- greatly accelerated junctional ectopic
Care Med 2003; 31:1082–1087 induced hypertension. Am J Physiol Renal tachycardia. J Am Coll Cardiol 1987; 10:
206. Weisinger JR, Bellorín-Font E: Magnesium Physiol 2006; 290:F1472–F1477 1095–1099
and phosphorus. Lancet 1998; 352:391–396 221. Sun Z, Zhang Z, Cade R: Renal responses to 234. Balaji S, Sullivan J, Deanfield J, et al: Mod-
207. Fischer UM, Cox CS Jr, Laine GA, et al: Mild chronic cold exposure. Can J Physiol Phar- erate hypothermia in the management of
hypothermia impairs left ventricular dia- macol 2003; 81:22–27 resistant automatic tachycardias in chil-
stolic but not systolic function. J Invest 222. Moat NE, Lamb RK, Edwards JC, et al: In- dren. Br Heart J 1991; 66:221–224
Surg 2005; 18:291–296 duced hypothermia in the management of 235. Pfammatter JP, Paul T, Ziemer G, et al:
208. Goldberg LI: Effects of hypothermia on con- refractory low cardiac output states follow- Successful management of junctional
tractility of the intact dog heart. Am J ing cardiac surgery in infants and children. tachycardia by hypothermia after cardiac
Physiol 1958; 194:92–98 Eur J Cardiothorac Surg 1992; 6:579 –584 operations in infants. Ann Thorac Surg
209. Suga H, Goto Y, Igarashi Y, et al: Cardiac 223. Deakin CD, Knight H, Edwards JC, et al: 1995; 60:556 –560
cooling increases Emax without affecting Induced hypothermia in the postoperative 236. Frank SM, Satitpunwaycha P, Bruce SR, et
relation between O2 consumption and sys- management of refractory cardiac failure al: Increased myocardial perfusion and sym-
tolic pressure-volume area in dog left ven- following paediatric cardiac surgery. Anaes- pathoadrenal activation during mild core
tricle. Circ Res 1988; 63:61–71 thesia 1998; 53:848 – 853 hypothermia in awake humans. Clin Sci
210. Mikane T, Araki J, Suzuki S, et al: O2 cost of 224. Dalrymple-Hay MJ, Deakin CD, Knight H, (Lond) 2003; 104:503–508
contractility but not of mechanical energy et al: Induced hypothermia as salvage treat- 237. Nabel EG, Ganz P, Gordon JB, et al: Dilation
increases with temperature in canine left ment for refractory cardiac failure following of normal and constriction of atheroscle-
paediatric cardiac surgery. Eur J Cardiotho-
ventricle. Am J Physiol 1999; 277:H65–H73 rotic coronary arteries caused by the cold
rac Surg 1999; 15:515–518
211. Lewis ME, Al-Khalidi AH, Townend JN, et pressor test. Circulation 1988; 77:43–52
225. Moriyama Y, Iguro Y, Shimokawa S, et al:
al: The effects of hypothermia on human 238. Schefold JC, Storm C, Joerres A, et al: Mild
Successful application of hypothermia com-
left ventricular contractile function during therapeutic hypothermia after cardiac ar-
bined with intra-aortic balloon pump sup-
cardiac surgery. J Am Coll Cardiol 2002; rest and the risk of bleeding in patients with
port to low-cardiac-output state after open
39:102–108 acute myocardial infarction. Int J Cardiol
heart surgery. Angiology 1996; 47:595–599
212. Rose JC, McDermott TF, Lilienfield LS, et 2008; 132:387–391
226. Yahagi N, Kumon K, Watanabe Y, et al:
al: Cardiovascular function in hypothermic 239. Tortorici MA, Kochanek PM, Poloyac SM:
Value of mild hypothermia in patients who
anesthetized man. Circulation 1957; 15: Effects of hypothermia on drug disposition,
have severe circulatory insufficiency even
512–517 metabolism, and response: A focus of hypo-
after intra-aortic balloon pump. J Clin
213. Thoresen M, Whitelaw A: Cardiovascular thermia-mediated alterations on the cyto-
Anesth 1998; 10:120 –125
changes during mild therapeutic hypother- chrome P450 enzyme system. Crit Care
227. Hovdenes J, Laake JH, Aaberge L, et al:
mia and rewarming in infants with Therapeutic hypothermia after out-of- Med 2007; 35:2196 –2204
hypoxic–ischemic encephalopathy. Pediat- hospital cardiac arrest: Experiences with 240. Kurz A, Sessler DI, Lenhardt R, et al: Peri-
rics 2000; 106:92–99 patients treated with percutaneous coro- operative normothermia to reduce the inci-
214. Mattheussen M, Mubagwa K, Van Aken H, et nary intervention and cardiogenic shock. dence of surgical-wound infection and
al: Interaction of heart rate and hypother- Acta Anaesthesiol Scand 2007; 51:137–142 shorten hospitalization. N Engl J Med 1996;
mia on global myocardial contraction of the 228. Skulec R, Kovarnik T, Dostalova G, et al: 334:1209 –1215
isolated rabbit heart. Anesth Analg 1996; Induction of mild hypothermia in cardiac 241. Peterson K, Carson S, Carney N: Hypother-
82:975–981 arrest survivors presenting with cardio- mia treatment for traumatic brain injury: A
215. Tveita T, Ytrehus K, Myhre ES, et al: Left genic shock syndrome. Acta Anaesthesiol systematic review and meta-analysis. J Neu-
ventricular dysfunction following rewarm- Scand 2008; 52:188 –194 rotrauma 2008; 25:62–71
ing from experimental hypothermia. J Appl 229. Oddo M, Ribordy V, Feihl F, et al: Early 242. Schwab S, Georgiadis D, Berrouschot J, et
Physiol 1998; 85:2135–2139 predictors of outcome in comatose survi- al: High risk of pneumonia in stroke pa-
216. Polderman KH, Tjong Tjin Joe R, Peerde- vors of ventricular fibrillation and non- tients under hypothermia. Feasibility and
man SM, et al: Effects of artificially induced ventricular fibrillation cardiac arrest safety of moderate hypothermia after mas-
hypothermia on intracranial pressure and treated with hypothermia: A prospective sive hemispheric infarction. Stroke 2001;
outcome in patients with severe traumatic study. Crit Care Med 2008; 36:2296 –2301 32:2033–2035
head injury. Intensive Care Med 2002; 28: 230. Boddicker KA, Zhang Y, Zimmerman MB, et 243. De Jonge E, Schultz MJ, Spanjaard L, et al:
1563–1567 al: Hypothermia improves defibrillation Effects of selective decontamination of di-
217. Pozos RS, Danzl D: Human physiological success and resuscitation outcomes from gestive tract on mortality and acquisition of
responses to cold stress and hypothermia. ventricular fibrillation. Circulation 2005; resistant bacteria in intensive care: A ran-
In: Medical Aspects of Harsh Environments. 111:3195–3201 domised controlled trial. Lancet 2003; 362:
Vol. 1. Textbooks of Military Medicine. Pan- 231. Rhee BJ, Zhang Y, Boddicker KA, et al: 1011–1016
dolf KB and Burr RE (Eds). Washington, Effect of hypothermia on transthoracic de- 244. De Smet AM, Kluytmans JA, Cooper BS, et
DC, Walter Reed Army Medical Center Bor- fibrillation in a swine model. Resuscitation al: Decontamination of the digestive tract
den Institute, 2001, pp 351–382 2005; 65:79 – 85 and oropharynx in intensive care patients.
218. Morgan ML, Anderson RJ, Ellis MA, et al: 232. Harada M, Honjo H, Yamazaki M, et al: New Engl J Med 2009; 360:20 –31

S202 Crit Care Med 2009 Vol. 37, No. 7 (Suppl.)

Anda mungkin juga menyukai