Consequences of Perioperative Hypothermia

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Handbook of Clinical Neurology, Vol.

157 (3rd series)


Thermoregulation: From Basic Neuroscience to Clinical Neurology, Part II
A.A. Romanovsky, Editor
https://doi.org/10.1016/B978-0-444-64074-1.00041-0
Copyright © 2018 Elsevier B.V. All rights reserved

Chapter 41

Consequences of perioperative hypothermia


KURT RUETZLER AND ANDREA KURZ*
Departments of General Anesthesiology and Outcomes Research, Anesthesiology Institute, Cleveland Clinic,
Cleveland, OH, United States

Abstract
Perioperative hypothermia is common, with an incidence ranging between 20 and 70%, and is defined
by a body core temperature below 36.0°C. Perioperative warming was rare during the previous century,
but was subsequently identified as a significant contributor to perioperative morbidity and mortality.
Perioperative hypothermia causes impaired pharmacodynamics, surgical site infections, blood loss and
coagulopathy, transfusion requirements, thermal discomfort, prolonged recovery, and prolonged duration
of hospitalization.
Measurement of central core temperature, maintaining normothermia, and consequent warming of
patients in the perioperative period are therefore essential. Several warming devices are commercially
available, including active skin warming as the most efficient, inexpensive, easy-to-use and mostly having
a good cost/benefit ratio for the majority of patients and surgeries.

Perioperative hypothermia is broadly defined as a body Scott et al., 2015). At any given time of day, core temp-
core temperature of less than 36.0oC. Induction of anes- erature is actively controlled by the thermoregulatory
thesia causes several physiologic changes. In particular, system (Tayefeh et al., 1998). In other words, circadian
the so-called redistribution effect within 60–90 minutes temperature changes are not passive responses to envi-
of anesthesia causes a decrease in body core temperature. ronmental perturbations – which suggests that humans
The incidence of perioperative hypothermia has been function best at temperatures near 37°C.
reported to range between 20 and 70% of patients having
surgery (Forstot, 1995; Hart et al., 2011).
PERIOPERATIVE HYPOTHERMIA
Most of the thermoregulation studies were performed
IS COMMON AFTER INDUCTION OF
in the 1990s, when patient warming was rare and scien-
ANESTHESIA, BUT NOT AT THE END
tific evidence about the consequences of perioperative
OF SURGERY
hypothermia was weak. However, contemporary scien-
tific evidence is mostly based on these “old” studies. Figure 41.1 displays the distribution of core temperature
One major limitation of several studies is that authors as a function of time after induction of anesthesia in a
previously defined perioperative normothermia as core cohort of 58,814 patients having noncardiac surgery at
temperature 36°C. The difficulty is that 36°C is never the Cleveland Clinic (Sun et al., 2015). During the first
a normal temperature in humans. Even at the circadian hour of anesthesia, core temperature typically drops
nadir, usually about 3.00 a.m., core temperature is not about 1°C, resulting in a median core temperature of
normally below 36.5°C; and at about 3.00 p.m., core about 35.8°C. The rapid drop of core temperature after
temperature is typically about 37.5°C (Sessler et al., induction of anesthesia is caused by balancing the
1991). On average, then, normal body temperature in temperature between the “cold” peripheral thermal com-
humans is about 37°C, not the 36°C that is widely accepted partment and the “warm” core thermal compartment.
as suitable for perioperative patients (Castren et al., 2009; This period is therefore a shift of heat from the warm

*Correspondence to: Andrea Kurz, Anesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland OH 44195,
United States. E-mail: ak@or.org
688 K. RUETZLER AND A. KURZ
38
Core
60 Temperature
Core Temperature (°C)

Core
Temperature Threshold
37 Quantile <36.0°C
<35.5°C
Top 5% 50

Incidence of Hypothermia
<35.0°C
Top 10%
Q3
36
Median
Q1
40
Bottom 10%
35 Bottom 5%
30

0 1 2 3 4 5 6
Time After Induction (h) 20
Sample Size

60k
30k
10
0k

Fig. 41.1. Distribution of core temperature as a function of


0
time after induction. (Reproduced from Sun Z, Honar H,
Sessler DI, et al. (2015) Intraoperative core temperature pat- 0 1 2 3 4 5 6
terns, transfusion requirement, and hospital duration in patients Time After Induction (h)
warmed with forced air. Anesthesiology 122: 276-285.) Fig. 41.2. Incidence of hypothermia as a function of time after
induction, under progressive core temperature thresholds defin-
core to the cold peripheral thermal compartment and is ing hypothermia. (Reproduced from Sun Z, Honar H, Sessler
DI, et al. (2015) Intraoperative core temperature patterns, trans-
accelerated by the vasodilatation caused by anesthesia.
fusion requirement, and hospital duration in patients warmed
Therefore, this period is called redistribution. This des- with forced air. Anesthesiology 122: 276-285.)
ignation also makes clear that the actual amount of heat
is not decreased (or at least, minimally) during this
period; the overall amount of heat is just distributed
elsewhere (Chapter 37). 60
However, core temperature increased after the initial
Percent of Patients

Episode
drop (as patients were actively warmed!), resulting in Duration
normothermia in the overwhelming majority of all 40 >15 minutes
>30 minutes
patients at the end of surgery. Generally, perioperative >60 minutes
>90 minutes
hypothermia is common in the first hours after induction >120 minutes
of anesthesia, and is it is therefore clinically essential to 20 >180 minutes
>240 minutes
counteract this.
Figure 41.2 displays the incidence of hypothermia
0
under various core temperature thresholds. After
34.0 34.5 35.0 35.5 36.0
45 minutes of induction of general anesthesia, almost Threshold Defining Hypothermia
two-thirds of patients reached the hypothermia threshold
Fig. 41.3. Incidence of (any) hypothermic episodes during the
of 36°C. In addition, more than one-quarter reached
case, according to progressive core temperature thresholds
the threshold of 35.5°C within 51 minutes of induction. defining hypothermia. (Reproduced from Sun Z, Honar H,
After 71 minutes, 7% of all patients even reached 35°C. Sessler DI, et al. (2015) Intraoperative core temperature pat-
Although perioperative hypothermia is most common terns, transfusion requirement, and hospital duration in patients
in the first hour after induction of anesthesia, it neverthe- warmed with forced air. Anesthesiology 122: 276-285.)
less remains common in about 20% of all patients, even
after 6 hours of surgery.
The duration of perioperative hypothermia varies. As a consequence, perioperative hypothermia signif-
Nearly half of all patients have core temperature < 36°C, icantly contributes to perioperative morbidity and mor-
and about 20 % of all patients have a core temperature tality (Chapter 33). Specifically, several randomized
< 35.5°C for more than 1 hour (Fig. 41.3). trials have shown that mild perioperative hypothermia
Most cellular functions and enzymatic activities causes impaired pharmacodynamics, surgical site infec-
are temperature-dependent. Hypothermia also provokes tions (SSIs), blood loss and coagulopathy, transfusion
systemic responses, some of which are potentially harm- requirements, thermal discomfort, prolonged recovery,
ful for the hypothermic patient. and prolonged duration of hospitalization.
CONSEQUENCES OF PERIOPERATIVE HYPOTHERMIA 689
IMPAIRED PHARMACODYNAMICS concentration in the cells. Hypothermia increases the
solubility of volatile anesthetics but does not appear
Most cellular functions and enzyme activity are
to alter the potency (Reynolds et al., 2008).
temperature-dependent. Therefore, it is unsurprising
However, with volatile anesthetics, the minimum
that even mild hypothermia prolongs the actions of var-
alveolar concentration (MAC) is diminished, and thus
ious drugs. By affecting drug metabolism, perioperative
a lower amount of agent is needed to prevent a reac-
hypothermia is associated with delayed emergence from
tion due to surgical stimulus. The MAC of halothane
anesthesia and prolonged stay in the postanesthesia care
and isoflurane decreases by approximately 5%/1oC as
unit (PACU).
core temperature decreases (Eger and Johnson, 1987).
Simultaneously, a decrease of 5.1%/1oC in isoflurane
Propofol and fentanyl MAC is observed for every 1oC reduction of core temp-
Propofol is one of the most commonly used intravenous erature in children (Liu et al., 2001).
anesthetics. A 3oC decrease in core temperature results
in an approximately 28% increase in plasma propofol
concentration, largely as a result of reduced hepatic SURGICAL SITE INFECTIONS AND
blood flow. However, mild hypothermia does not reduce COMPLICATIONS (WOUND HEALING
propofol requirement during craniotomy surgery (Leslie AND DEHISCENCE)
et al., 2002).
One study reported that the steady-state plasma SSIs are the third leading cause of nosocomial infections,
concentration of fentanyl is increased by 5%/ 1oC drop accounting for 14–16% of all hospital-acquired infec-
of core temperature (Fritz et al., 2005). tions, and are the leading cause of nosocomial infections
among surgical patients (Esnaola and Cole, 2011).
Considering all of the complications that may be trig-
Muscle relaxants gered by anesthesia, SSIs are likely the most significant
Nondepolarizing muscle relaxants are markedly affected cause of morbidity, even greater than all other anesthetic
by perioperative hypothermia, resulting in a prolonged complications combined.
duration of action. This is based on a wide range of Perioperative hypothermia affects host defense
reasons, including changes in volume of distribution, against contamination via at least three mechanisms.
altered local diffusion receptor affinity, changes in First, perioperative hypothermia triggers postoperative
pH at the neuromuscular junction, and the net effect of vasoconstriction with the goal of constraining meta-
cooling on the various components of neuromuscular bolic heat to the core and speeds rewarming. On the
transmission. other hand, vasoconstriction also reduces perfusion of
The duration of action of vecuronium is more than dou- wounded tissues. As a consequence, tissue oxygen par-
bled in patients experiencing perioperative hypothermia tial pressure drops (even when blood is fully saturated),
(Heier et al., 1991). Interestingly, when neostigmine is which is especially important, as oxidative killing by
used, as an antagonist of vecuronium, it does not appear neutrophils, the primary defense against bacterial con-
to be altered by mild hypothermia (Heier et al., 2002). tamination, mostly relies on molecular oxygen (Edwards
Perioperative hypothermia prolongs the duration of et al., 1984; Allen et al., 1997).
action of atracurium by approximately 60% in patients with Second, perioperative hypothermia reduces systemic
a 3oC decrease of core temperature (Leslie et al., 1995), immune activation, including T-cell-mediated antibody
although atracurium appears not to be temperature- production, and decreases motility of key cells, including
sensitive. macrophages (Jonsson et al., 1988).
Surprisingly, there is no difference in the recovery Third, vasoconstriction-induced tissue hypoxia impairs
index (time for 25–75% twitch recovery) for both atra- tissue healing and protein metabolism, which are impor-
curium and vecuronium during normo- and hypothermia. tant to prevent wound dehiscence and recontamination
The duration of action of another muscle relaxant, (Carli et al., 1989).
rocuronium, was reported to be prolonged in car- Perioperative hypothermia is associated with a signif-
diac patients undergoing hypothermic cardiac bypass icant increase of infections in patients undergoing chole-
(Smeulers et al., 1995). cystectomy (Hart et al., 2011; Madrid et al., 2016). In
a major study by Kurz et al. (1996) investigating patients
having colon resection, the incidence of SSIs was
Volatile anesthetics
threefold greater in hypothermic versus normothermic
Anesthesia potency is driven by the steady-state patients (Fig. 41.4; Table 41.1). As a consequence,
plasma partial pressure opposed to the actual anesthetic hospitalization was prolonged in hypothermic patients.
690 K. RUETZLER AND A. KURZ
SSIs increase postoperative hospitalization by an aver- Platelets
age of 4 days, resulting in an increased attributable cost
Although platelet counts are stable and are not affected
of US$8000–25,000 for each patient (Hart et al., 2011).
by temperature, perioperative hypothermia affects plate-
A recent review summarized that there was a sig-
let function. It is commonly suggested that inhibition of
nificant benefit of actively warmed patients over nonac-
intrinsic platelet function is not the cause of the coagu-
tively warmed patients in the incidence of SSIs and
lopathy. In fact, it mostly results from the reversible
complications (risk ratio 0.36, 95% confidence interval
impairment of platelet aggregation via reduced release
(CI) 0.2–0.66, p ¼ 0.0008) (Madrid et al., 2016).
of thromboxane A3, which is necessary for the formation
of an initial platelet plug (Valeri et al., 1992). Furthermore,
BLOOD LOSS AND COAGULOPATHY perioperative hypothermia has been reported to induce
Perioperative hypothermia causes coagulopathy and morphologic changes of the platelets, and enhances the
consequently increased blood loss by several routes. binding of platelets to fibrinogen through activation of
GbIIb-IIa receptors.
25 25
P = 0.001
P < 0.01
Hospital Duration (days)
20 20

Enzymes of the coagulation cascade


Infections (%)

15 15
In addition, perioperative hypothermia impairs the func-
10 10 tion of several enzymes of the coagulation cascade and
finally reduces clot formation. This is especially impor-
5 5
tant, as this is frequently missed in the clinical setting,
0 0
as coagulation laboratory studies including prothrombin
Hypothermic Normothermic Hypothermic Normothermic time and partial thromboplastin time are usually per-
34.7 ± 0.6 36.6 ± 0.5 34.7 ± 0.6 36.6 ± 0.5
formed at a temperature of 37oC, rather than the patient’s
Core Temperature (°C)
actual temperature (Rohrer and Natale, 1992). The
Fig. 41.4. Hypothermia, wound infection and duration of
identical testing will be prolonged, if the testing is
hospitalization. (Reproduced with permission from Kurz A,
Sessler DI, Lenhardt R (1996) Perioperative normothermia to done at the patient’s actual temperature, instead of at
reduce the incidence of surgical-wound infection and shorten 37oC. Several in vitro studies reported prolongation
hospitalization. Study of Wound Infection and Temperature of up to 10% in hypothermic versus normothermic
Group. N Engl J Med 334: 1209–1215. Copyright © tests. However, whether this prolongation is clinically
Massachusetts Medical Society.) relevant remains questionable.

Table 41.1
Postoperative findings in the two study groupsa

Normothermia Hypothermia
Variable (n ¼ 104) (n ¼ 96) p-value

All patients
Infection: number of patients (%) 6 (6) 18 (19) 0.009
ASEPSIS score 7  10 13  16 0.002
Collagen deposition (mg/cm) 328  135 254  114 0.04
Days to first solid food 5.6  2.5 6.5  2.0 0.006
Days to suture removal 9.8  2.9 10.9  1.9 0.002
Days of hospitalization 12.1  4.4 14.7  6.5 0.001
Uninfected patients
Number of patients 98 78
Days to first solid food 5.2  1.6 6.1  1.6 <0.001
Days to suture removal 9.6  2.6 10.6  1.6 0.003
Days of hospitalization 11.8  4.1 13.5  4.5 0.01

Reproduced with permission from Kurz A, Sessler DI, Lenhardt R (1996) Perioperative normothermia to reduce the incidence of surgical-wound
infection and shorten hospitalization. Study of Wound Infection and Temperature Group. N Engl J Med 334: 1209–1215. Copyright © Massachu-
setts Medical Society.
a
Plus and minus values are means  standard deviation.
CONSEQUENCES OF PERIOPERATIVE HYPOTHERMIA 691
Fibrinolysis Although there are many divergent results from
several retrospective studies, two randomized trials have
Generally, the fibrinolytic system regulates the balance
confirmed that a reduction of only approximately 0.5oC
between the formation of hemostatic plugs and restora-
core temperature increases blood loss by 200–300 mL in
tion of blood flow after clot formation (Reynolds et al.,
patients undergoing hip arthroplasty (Winkler et al., 2000).
2008). Excessive fibrinolysis predisposes patients to
A comprehensive meta-analysis of randomized
hemorrhage, while inadequate fibrinolysis predisposes
controlled trials by Rajagopalan et al. (2008) compared
patients to thrombosis.
normothermic patients with patients experiencing peri-
The most important structural element in formed
operative hypothermia (34–36oC). Fourteen studies were
clots is fibrin, but it is subject to degradation by plasmin,
included in order to assess the relationship between
which represents the activated from of plasminogen.
hypothermia and blood loss: mild hypothermia increased
Therefore, activation of plasminogen and conversion
blood loss by approximately 16% (Table 41.2).
to plasmin represent the core steps of the fibrinolytic
system and are mostly mediated by tissue-type plasmin-
TRANSFUSION REQUIREMENT
ogen. Findings of several studies suggest that fibrinolysis
is not affected by mild perioperative hypothermia, There is no linear relationship between blood loss and
while hyperthermia intensively affects fibrinolysis. transfusion requirements. It seems to be obvious that
In conclusion, this might result in the assumption that increased blood loss increases the individual need for
hypothermia-induced coagulopathy does not result from transfusion requirements. Blood transfusion was expected
excessive clot lysis. Data from thromboelastograms to be safe for decades (taking into account the risk of
suggest that hypothermia impairs clot formation, rather human immunodeficiency virus (HIV) and hepatitis C),
than facilitating clot degeneration. but blood transfusions are more toxic than previously

Table 41.2
Total blood loss meta-analysis and forest plot

Sample Normothermic Hypothermic Outcome


Size (N) (N) (H) (N/H)
Study N:H mean (sd) mean (sd) mean (95% Cl)

Schmied 30 : 30 1670 (320) 2150 (550) 0.79 (0.70, 0.88)


Winkler 75 : 75 1531(1055, 1746) 1678(1366, 1965) 0.90 (0.82, 1.00)
Widman 22 : 24 923 (410) 1068 (482) 0.87 (0.68, 1.11)
Persson 29 : 30 186 (145) 308 (257) 0.62 (0.43, 0.89)
Hofer 29 : 29 1497 (497) 2300 (788) 0.65 (0.55, 0.77)
Bock 20 : 20 635 (507) 1070 (803) 0.58 (0.38, 0.89)
Johansson 25 : 25 1047 (413) 1066 (441) 0.99 (0.80, 1.23)
Smith 31 : 30 423 (562) 159 (268) 3.14 (1.82, 5.42)
Frank 142 : 158 390 (834) 520 (754) 0.56 (0.43, 0.73)
Mason 32 : 32 111 (40) 157 (73) 0.73 (0.60, 0.89)
Casati 25 : 25 470 (170) 442 (216) 1.11 (0.89, 1.40)
Murat 26 : 25 160 (61) 161 (100) 1.09 (0.84, 1.43)
Hohn 43 : 73 660(230, 1870) 956(340, 5480) 0.69 (0.36, 1.34)
Nathan 73 : 71 569 (356) 666 (405) 0.85 (0.70, 1.02)

Summary 0.84 (0.74, 0.96)


Treatment effect P = 0.009

0.4 1.0 2.0 3.0 4.0 5.0 6.0


Favors Favors
Normothermic Hypothermic

From Rajagopalan S, Mascha E, Na J, et al. (2008) The effects of mild perioperative hypothermia on blood loss and transfusion requirement. Anes-
thesiology 108: 71–77, with permission from Wolters Kluwer Health.
Treatment effect is expressed as ratio of geometric means of blood loss for normothermic (N) versus hypothermic (H) patients. Results indicate an
estimated 16% (95% confidence interval (CI) 4%, 26%) lower average blood loss in normothermic versus hypothermic patients, p < 0.009.
692 K. RUETZLER AND A. KURZ
Table 41.3
Transfusion meta-analysis and forest plot

Study Normothermic Hypothermic Outcome


n/N (%) n/N (%) RR (95%Cl)

Schmied 1/30 (3%) 7/30 (23%) 0.14 (0.02, 1.09)


Winkler 29/75 (39%) 40/75 (53%) 0.73 (0.51, 1.03)
Widman 9/22 (41%) 11/24 (46%) 0.89 (0.46, 1.73)
Hofer 5/29 (17%) 11/29 (38%) 0.45 (0.18, 1.14)
Johansson 15/25 (60%) 13/25 (52%) 1.15 (0.7, 1.89)
Kurz 23/104 (22%) 34/96 (35%) 0.62 (0.4, 0.98)
Bock 3/20 (15%) 9/20 (45%) 0.33 (0.11, 1.05)
Hohn 17/43 (40%) 18/43 (42%) 0.94 (0.57, 1.57)
Nathan 23/73 (32%) 24/71 (34%) 0.93 (0.58, 1.49)
Smith 2/31 (6%) 1/30 (3%) 1.94 (0.19, 20.24)

Summary 0.78 (0.63, 0.97)


Treatment effect P = 0.027

0.1 1.0 2.0 3.0 4.0

Favors Favors
Normothermic Hypothermic

Reproduced from Rajagopalan S, Mascha E, Na J, et al. (2008) The effects of mild perioperative hypothermia on blood loss and transfusion require-
ment. Anesthesiology 108: 71–77.
Treatment effect is expressed as the relative risk (RR) of transfusion in normothermic versus hypothermic patients. Normothermia is associated with
22% less risk of transfusion than hypothermia (95% confidence interval (CI) 3%, 37%), p ¼ 0.027. n, number transfused; N. number of patients.

believed. Data from the above-mentionedmeta-analysis


by Rajagopalan et al. (2008) reported that mild hypother- Table 41.4
mia significantly increased the relative risk for transfusion Hypothermia and transfusion requirement
by approximately 22% (CI 3–37%) (Table 41.3).
In a retrospective study performed at the Cleveland Adjusteda odds ratio Adjusteda ratio of
Clinic, about 4.6% of all patients received a blood trans- (pointwise 95% CI) geometric mean
fusion (Sun et al., 2015). Based on a multivariate logistic Area under for intraoperative duration of
regression model, the authors found a significant asso- 37°C erythrocyte hospitalization
ciation between core temperature <37oC and risk of hav- (degree transfusion (pointwise 95% CI)
hours) (n ¼ 45,866) (n ¼ 39,180)
ing a transfusion. The odds ratios relative to a reference
value of 1oC are presented in Table 41.4. 0.25 1.34 (1.04, 1.73) 0.96 (0.90, 1.03)
Generally, risk of transfusion significantly increases 0.50 1.10 (0.98, 1.24) 0.97 (0.93, 1.01)
as a function of duration and dimension of perioperative 1.00 (Reference) (Reference)
hypothermia. 2.00 1.00 (0.89, 1.13) 1.03 (1.00, 1.05)
Figure 41.5 displays the adjusted probability of trans- 4.00 1.12 (0.91, 1.39) 1.06 (1.03, 1.09)
fusion estimates versus integrated area above the core 8.00 1.41 (1.08, 1.84) 1.05 (1.01, 1.10)
temperature versus time curve and <37oC. The shaded 16.00 2.02 (1.30, 3.14) 0.96 (0.86, 1.08)
regions represent pointwise 95% CI.
Reproduced from Sun Z, Honar H, Sessler DI, et al. (2015) Intraoperative
core temperature patterns, transfusion requirement, and hospital dura-
SHIVERING tion in patients warmed with forced air. Anesthesiology 122: 276–285.
a
Estimates adjusted for year, type, and duration of surgery, body mass
Shivering is an autonomic thermoregulatory response,
index, age, preoperative platelet count, preoperative hemoglobin,
defined as rhythmic involuntary muscular activity. estimated blood loss, and individual anesthesiologist, as well as the
The ultimate goal of shivering is to produce heat in order Elixhauser comorbidities, which are listed in Table 41.2.
to restore normothermia. CI, confidence interval.
CONSEQUENCES OF PERIOPERATIVE HYPOTHERMIA 693
35%
experienced cold during the postoperative period as the
worst part of their hospitalization, sometimes even worse
than surgical pain. Thermal discomfort is also stressful
30%
for patients, and has been reported to lead to higher
blood pressure, heart rate, and plasma catecholamine
Adjusted Pr(Transfusion)*

25%
concentrations (Frank et al., 1995b). Active warming
improves thermal comfort in hypothermic patients, but
20%
prevention of postoperative hypothermia is obviously
the preferable management strategy.
15%

10% CARDIAC COMPLICATIONS


Among the major complications of hypothermia, myo-
5% cardial outcomes are least well established. An observa-
tional analysis suggests an association between Surgical
0% Care Improvement Project-10 (SCIP-10) compliance and
1/4 1/2 1 2 4 8 16
temperature and cardiovascular outcomes (Scott et al.,
Area Under 37°C (degree⋅hours) 2015). A single-center randomized trial of 300 patients
Fig. 41.5. Hypothermia and transfusion requirement. LOS,
evaluated cardiac outcomes, although the study was seri-
length of stay. (Reproduced from Sun Z, Honar H, Sessler ously underpowered (Frank et al., 1997). For example,
DI, et al. (2015) Intraoperative core temperature patterns, only two hypothermic patients had a cardiac arrest
transfusion requirement, and hospital duration in patients and one had a myocardial infarction versus none in the
warmed with forced air. Anesthesiology 122: 276-285.) normothermic group (36.7°C). Ten hypothermic patients
experienced cardiovascular events versus two normother-
mic patients. These fragile results, based on fewer than
Intraoperative shivering is rare, as many patients 10 outcome events, are nearly as likely to be wrong as
receive muscle relaxants and the shivering threshold is to be right (Ioannidis, 2005), and are a poor basis for health
significantly reduced by anesthetic agents and only policy. The other relevant study had only 100 patients
patients having serious hypothermia hit this threshold. and was thus even more fragile (Elmore et al., 1998).
In contrast, postoperative shivering in hypother- An additional limitation of the study by Frank et al.
mic patients is common. Shivering is largely thermo- (1997) is that diagnosis was primarily based on Holter
regulatory, in order to produce heat and compensate for electrocardiogram finding rather than troponin concentra-
hypothermia, but is aggravated by volatile anesthetics. tions, which are much more sensitive. Consequently, the
However, thermoregulatory major muscular activity must overall myocardial infarction rate was <1%, whereas the
be distinguished from low-intensity shivering-like mus- true rate is no less than 10% in vascular surgery patients.
cular activity, which is common in patients in pain It is highly questionable whether any conclusion about
(Horn et al., 1999). On average, postoperative shivering hypothermia and myocardial outcomes can be derived
increases oxygen consumption by approximately 40% from a study that missed 90% of the presumed myocardial
(Frank et al., 1995a). It appears to be logical that this events. The only other relevant trial was restricted to 100
increase of oxygen consumption is associated with peri- patients, leaving it even less powered to detect clinically
operative morbidity and mortality. Shivering is extremely important outcomes (Elmore et al., 1998). There thus
uncomfortable for patients and is a preventable complica- remains considerable doubt as to the true effect of
tion of perioperative hypothermia. However, shivering moderate hypothermia on cardiovascular outcomes.
can be treated effectively by a variety of medications,
including pethidine, clonidine, dexmedetomidine, and DELAYED DISCHARGE FROM THE PACU
ketamine. Lowering the shivering threshold is the
assumed mechanism. It may not be not very surprising that perioperative hypo-
thermia is associated with delayed discharge from the
PACU. In a mayor randomized trial, Lenhardt et al.
THERMAL DISCOMFORT AND ANXIETY
(1997) assigned patients undergoing elective surgery to
Thermal discomfort is a predictable and common conse- normothermia or hypothermia during anesthesia. The
quence of patients experiencing perioperative hypother- authors assessed length of stay in the PACU until a sus-
mia. Even mild hypothermia produces marked thermal tained score of 13 was reached. Ability to discharge was
discomfort and consequently affects patient satisfaction based on modification of the Aldrete and Kroulik (1970)
(Reynolds et al., 2008). Patients often identify having scoring system. The score is based on activity, ventilation,
694 K. RUETZLER AND A. KURZ
consciousness, and hemodynamic responses; 0, 1, or 2 higher risk of experiencing an SSI. It seems to be obvious
points were assigned for each of eight responses. In this that corresponding hospitalization in patients having an
study, hypothermic patients required about 40 minutes SSI is prolonged. Interestingly, the important effect of
longer than normothermic patients to reach a sustained prolongation of hospitalization was present in both, all
score 13, the defined criterion for discharge. Core (12.1  4.4 vs. 14.7  6.5 days), and in infected (11.8
temperature in the hypothermic patients required 134  4.1 vs. 13.5  4.5 days) patients. Therefore, perioper-
 60 minutes to reach 36°C. Consequently, hypothermic ative hypothermia per se (as well as the higher risk of
patients required about 90 minutes longer than normother- having an SSI) prolongs hospitalization (Table 41.1).
mic patients to reach both a sustained score  13 and a core As is consistent with a delay in clinical healing,
temperature >36°C (p < 0.001) (Figs 41.6 and 41.7) sutures were removed significantly later and the deposi-
(Lenhardt et al., 1997). tion of collagen (an index of scar formation and the
strength of the healing wound) was significantly less in
the hypothermia group than in the normothermia group.
PROLONGED HOSPITALIZATION
That the patients assigned to hypothermia required
Two hundred patients having elective colorectal resec- significantly more time before they could tolerate solid
tion for cancer or inflammatory bowel disease were food is also consistent with impaired healing.
randomly assigned to normothermia (37  0.3°C) vs. In another study consisting of 39,180 patients, Sun
hypothermia (34.4  0.4°C) in a major landmark study et al. (2015) reported a significant association between
by Kurz et al. (1996). Hypothermia caused a threefold area <37oC core temperature threshold and geometric
mean duration of hospitalization (Fig. 41.8).
Consequently, prolongation of hospitalization seems
to be essential, especially as healthcare-related costs tend
to rise sharply. Even extremely cautious assessments of
costs per in-hospital day of US$1000 represent increased
overall costs of about US$2000 in every single patient
experiencing perioperative hypothermia.

3.0

Fig. 41.6. Kaplan–Meier “survival” analysis showing the


percentage of patients not sustaining a recovery score 13.
Adjusted Geometric Mean LOS*

2.8
The probability value, using a Wilcoxon analysis, was < 0.0001.
(Reproduced from Lenhardt R, Marker E, Goll V, et al. (1997)
Mild intraoperative hypothermia prolongs postanesthetic 2.6
recovery. Anesthesiology 87: 1318–1323.)

2.4

2.2

2.0

1/4 1/2 1 2 4 8 16
Area Under 37°C (degree⋅hours)
Fig. 41.8. Estimates of geometric mean duration of hospital-
Fig. 41.7. Kaplan–Meier “survival” analysis showing the ization in days vs. integrated area above the core temperature
percentage of patients not sustaining a recovery score 13 and vs. time curve and <37oC core temperature. The shaded
a core temperature 36°C. The probability value, using a Wil- regions represent pointwise 95% confidence interval. LOS,
coxon analysis, was 0.0001.34 (Reproduced from Lenhardt length of stay. (Reproduced from Sun Z, Honar H, Sessler
R, Marker E, Goll V, et al. (1997) Mild intraoperative hypo- DI, et al. (2015) Intraoperative core temperature patterns,
thermia prolongs postanesthetic recovery. Anesthesiology 87: transfusion requirement, and hospital duration in patients
1318–1323.) warmed with forced air. Anesthesiology 122: 276-285.)
CONSEQUENCES OF PERIOPERATIVE HYPOTHERMIA 695
STRATEGIES FOR MAINTAINING 2. Active skin warming: Convective warming of the
NORMOTHERMIA DURING skin with forced air is by far the most frequently used
ANESTHESIA and effective perioperative warming device. Forced-
air warming is especially attractive, as it is easy to
Measurement of central core temperature use, inexpensive, and for most patients and surgery
It is essential to focus on central core temperature, as the has a good cost/benefit ratio. In order to achieve the
core thermal component is usually relatively well and highest effectiveness, the forced-air blankets should
homogeneously perfused. In contrast, the peripheral be directly placed on the patient’s skin and should
thermal component is less perfused and consequently cover as much of the body as possible. An alternative
relatively hypothermic compared to the central thermal approach is conductive warming using resistive
component. Therefore, measuring the central core tem- heating or circulating water (Ruetzler et al., 2011;
perature is essential and rewarming should be guided Hasegawa et al., 2012).
by central core temperature alone. The central core (a) Fluid warming is not efficient, as the tempera-
temperature can be reliably measured at four sites: ture of the infusion (usually not warmer than
38–39°C) can only slightly exceed core temper-
1. pulmonary artery (using a Swan catheter)
ature. Consequently, active warming of the
2. distal esophagus
patient is basically impossible unless very large
3. nasopharynx with the probe inserted 10–20 cm
amounts of fluid are given over a short period of
4. tympanic membrane, if measured with a contact
time. The most important effect of fluid warm-
thermistor or thermocouple.
ing is decreasing the amount of heat loss. For
The temperatures measured at these four sites do not example, 1 liter of crystalloid given at room
vary more than a couple of tenths of 1°C and therefore temperature decreases core temperature by
even a single measurement gives a reliable estimation 0.25°C. Equally 1 unit of blood from the refrig-
of the actual core temperature. erator decreases core temperature by 0.25°C.
(b) Warming inspired and peritoneal gases: warm-
Prewarming ing patients by warming inhaled air is insuffi-
cient, as the heat capacity of air is low. Only a
Core temperature drops during the initial hour of anesthe- marginal amount of heat is lost via the respira-
sia based on the principle of redistribution (see above). tory system. Thus warmed air is unable to trans-
The redistribution effect might be partially attenuated fer a considerable amount of heat to patients
by prewarming patients. It is important to understand that (Sammour et al., 2010).
prewarming does not increase patients’ core temperature, (c) Vascular heat exchange catheters transfer much
but increases the temperature of the peripheral tissues and more heat compared to skin-warming devices.
therefore decreases the amount of redistribution hypother- On the other hand, heat exchange catheters
mia. Consequently, prewarming alone does not prevent are quite expensive, and placement is invasive.
perioperative hypothermia, but it does reduce the drop Thus, their use is mostly restricted to patients
in temperature during the initial period of anesthesia. Typ- requiring a rapid onset of hypothermia, for
ically, the core temperature in prewarmed patients stays example after distinct accidental hypothermia
about 0.4°C warmer compared to nonprewarmed patients. (Sessler, 2016).

Perioperative warming devices


Various perioperative warming devices are available and SUMMARY
can be divided into passive insulation and active warm-
Perioperative hypothermia is common, and significantly
ing devices of the skin surface, fluids, inspired and peri-
affects perioperative morbidity and mortality. In particu-
toneal gases, and endovascular heat exchangers.
lar, perioperative hypothermia causes impaired pharmaco-
1. Passive insulation: a single layer of passive insulation dynamics, SSIs, blood loss and coagulopathy, transfusion
reduces cutaneous heat loss by 30%. However, it is requirements, thermal discomfort, prolonged recovery,
essential to determine that the layer does not actively and prolonged duration of hospitalization.
transfer heat into the body, even by adding several Maintenance or restoration of normothermia is essen-
extra layers of insulation (Sessler and Schroeder, tial in all surgical patients and can effectively be achieved
1993). Consequently, passive insulation can decrease by consequent (re-)warming during surgery. Core temper-
heat loss but will not add any benefit in the mainte- ature should be measured in all patients having general or
nance of perioperative normothermia (Sessler, 2016). regional anesthesia lasting more than 30 minutes.
696 K. RUETZLER AND A. KURZ
Important points Hasegawa K, Negishi C, Nakagawa F et al. (2012). Core
temperatures during major abdominal surgery in patients
● Perioperative hypothermia is common. warmed with new circulating-water garment, forced-air
● Perioperative hypothermia is a significant contri- warming, or carbon-fiber resistive-heating system. Journal
butor to morbidity and mortality. of Anesthesia 26: 168–173.
● Maintenance and restoration of normothermia are Heier T, Caldwell JE, Sessler DI et al. (1991). Mild intraopera-
essential. tive hypothermia increases duration of action and sponta-
● Monitoring the core temperature during surgery and neous recovery of vecuronium blockade during nitrous
consequent warming of the patients are imperative. oxide-isoflurane anesthesia in humans. Anesthesiology
74: 815–819.
Heier T, Clough D, Wright PM et al. (2002). The influence of
REFERENCES mild hypothermia on the pharmacokinetics and time course
of action of neostigmine in anesthetized volunteers.
Aldrete JA, Kroulik D (1970). A postanesthetic recovery Anesthesiology 97: 90–95.
score. Anesth Analg 49: 924–934. Horn EP, Schroeder F, Wilhelm S et al. (1999).
Allen DB, Maguire JJ, Mahdavian M et al. (1997). Wound Postoperative pain facilitates nonthermoregulatory tremor.
hypoxia and acidosis limit neutrophil bacterial killing Anesthesiology 91: 979–984.
mechanisms. Arch Surg 132: 991–996. Ioannidis JP (2005). Contradicted and initially stronger effects
Carli F, Emery PW, Freemantle CA (1989). Effect of peropera- in highly cited clinical research. JAMA 294: 218–228.
tive normothermia on postoperative protein metabolism in Jonsson K, Hunt TK, Mathes SJ (1988). Oxygen as an isolated
elderly patients undergoing hip arthroplasty. Br J Anaesth variable influences resistance to infection. Ann Surg 208:
63: 276–282. 783–787.
Castren M, Silfvast T, Rubertsson S et al. (2009). Scandinavian Kurz A, Sessler DI, Lenhardt R (1996). Perioperative normo-
clinical practice guidelines for therapeutic hypothermia thermia to reduce the incidence of surgical-wound infection
and post-resuscitation care after cardiac arrest. Acta and shorten hospitalization. Study of Wound Infection and
Anaesthesiol Scand 53: 280–288. Temperature Group. N Engl J Med 334: 1209–1215.
Edwards SW, Hallett MB, Campbell AK (1984). Oxygen- Lenhardt R, Marker E, Goll V et al. (1997). Mild intra-
radical production during inflammation may be limited operative hypothermia prolongs postanesthetic recovery.
by oxygen concentration. Biochem J 217: 851–854. Anesthesiology 87: 1318–1323.
Eger 2nd EI, Johnson BH (1987). MAC of I-653 in rats, includ- Leslie K, Sessler DI, Bjorksten AR et al. (1995). Mild hypo-
ing a test of the effect of body temperature and anesthetic thermia alters propofol pharmacokinetics and increases
duration. Anesth Analg 66: 974–976. the duration of action of atracurium. Anesth Analg 80:
Elmore JR, Franklin DP, Youkey JR et al. (1998). 1007–1014.
Normothermia is protective during infrarenal aortic surgery. Leslie K, Bjorksten AR, Ugoni A et al. (2002). Mild core hypo-
J Vasc Surg 28: 984–992. thermia and anesthetic requirement for loss of responsive-
Esnaola NF, Cole DJ (2011). Perioperative normothermia ness during propofol anesthesia for craniotomy. Anesth
during major surgery: is it important? Adv Surg 45: Analg 94: 1298–1303. table of contents.
249–263. Liu M, Hu X, Liu J (2001). The effect of hypothermia
Forstot RM (1995). The etiology and management of inadver- on isoflurane MAC in children. Anesthesiology 94:
tent perioperative hypothermia. J Clin Anesth 7: 657–674. 429–432.
Frank SM, Fleisher LA, Olson KF et al. (1995a). Multivariate Madrid E, Urrutia G, Roque i Figuls M et al. (2016). Active
determinants of early postoperative oxygen consumption in body surface warming systems for preventing complica-
elderly patients. Effects of shivering, body temperature, tions caused by inadvertent perioperative hypothermia in
and gender. Anesthesiology 83: 241–249. adults. Cochrane Database Syst Rev 4CD009016.
Frank SM, Higgins MS, Breslow MJ et al. (1995b). The cate- Rajagopalan S, Mascha E, Na J et al. (2008). The effects of
cholamine, cortisol, and hemodynamic responses to mild mild perioperative hypothermia on blood loss and transfu-
perioperative hypothermia. A randomized clinical trial. sion requirement. Anesthesiology 108: 71–77.
Anesthesiology 82: 83–93. Reynolds L, Beckmann J, Kurz A (2008). Perioperative
Frank SM, Fleisher LA, Breslow MJ et al. (1997). complications of hypothermia. Best Pract Res Clin
Perioperative maintenance of normothermia reduces the Anaesthesiol 22: 645–657.
incidence of morbid cardiac events: a randomized clinical Rohrer MJ, Natale AM (1992). Effect of hypothermia on the
trial. JAMA 277: 1127–1134. coagulation cascade. Crit Care Med 20: 1402–1405.
Fritz HG, Holzmayr M, Walter B et al. (2005). The effect of Ruetzler K, Kovaci B, Guloglu E et al. (2011). Forced-air and a
mild hypothermia on plasma fentanyl concentration and novel patient-warming system (vitalHEAT vH2) compara-
biotransformation in juvenile pigs. Anesth Analg 100: bly maintain normothermia during open abdominal sur-
996–1002. gery. Anesthesia and Analgesia 112: 608–614.
Hart SR, Bordes B, Hart J et al. (2011). Unintended perioper- Sammour T, Kahokehr A, Hayes J et al. (2010). Warming
ative hypothermia. Ochsner J 11: 259–270. and humidification of insufflation carbon dioxide in
CONSEQUENCES OF PERIOPERATIVE HYPOTHERMIA 697
laparoscopic colonic surgery: a double-blinded random- pharmacodynamics and pharmacokinetics of rocuronium.
ized controlled trial. Ann Surg 251: 1024–1033. J Cardiothorac Vasc Anesth 9: 700–705.
Scott AV, Stonemetz JL, Wasey JO et al. (2015). Compliance Sun Z, Honar H, Sessler DI et al. (2015). Intraoperative
with Surgical Care Improvement Project for body tempera- core temperature patterns, transfusion requirement, and
ture management (SCIP Inf-10) is associated with improved hospital duration in patients warmed with forced air.
clinical outcomes. Anesthesiology 123: 116–125. Anesthesiology 122: 276–285.
Sessler DI (2016). Perioperative thermoregulation and heat Tayefeh F, Plattner O, Sessler DI et al. (1998). Circadian
balance. Lancet 387: 2655–2664. changes in the sweating-to-vasoconstriction interthreshold
Sessler DI, Schroeder M (1993). Heat loss in humans range. Pflugers Arch 435: 402–406.
covered with cotton hospital blankets. Anesth Analg 77: Valeri CR, Khabbaz K, Khuri SF et al. (1992). Effect of skin
73–77. temperature on platelet function in patients undergoing
Sessler DI, Lee KA, McGuire J (1991). Isoflurane anesthesia extracorporeal bypass. J Thorac Cardiovasc Surg 104:
and circadian temperature cycles. Anesthesiology 75: 108–116.
985–989. Winkler M, Akca O, Birkenberg B et al. (2000). Aggressive
Smeulers NJ, Wierda JM, van den Broek L et al. (1995). warming reduces blood loss during hip arthroplasty.
Effects of hypothermic cardiopulmonary bypass on the Anesth Analg 91: 978–984.

You might also like