You are on page 1of 9

A d v a n c e s i n An e s t h e s i a

Monitoring
Yi Deng, MDa,*, Jovany Cruz Navarro, MDb,c, Sandeep Markan, MDa

KEYWORDS
 Anesthesia monitoring  Perioperative monitoring  Neurologic monitoring
 Cardiovascular monitoring

KEY POINTS
 There have been many advances in perioperative anesthetic monitoring, specifically in the fields of
neurologic and cardiovascular monitoring.
 Neurologic monitoring is necessary to prevent nerve injury, ensure adequate depth of anesthesia,
and optimize cerebral oxygenation.
 Minimally invasive cardiovascular monitors use dynamic measures of fluid responsiveness to better
assess and manage the patient’s volume status.

INTRODUCTION of neural function because of anesthesia compli-


cations during surgery is a major loss to the
Patient monitoring during the administration of individual. Advances during the last three decades
anesthesia is essential for safety and a core tenet have led to improvements in clinical neuromonitor-
of anesthesiology. The American Society of Anes- ing technology. The goal of neuromonitoring is to
thesiology established guidelines and promotes assess and preserve the functional integrity of
minimum practice standards of monitoring cardio- the brain, brainstem, spinal cord, and/or peripheral
pulmonary function and perfusion. These include nerves during surgery. If mechanical or physio-
heart rate and rhythm, blood pressure (BP), tem- logic injury to these structures is suspected,
perature, oxygenation, and ventilation. Adjunctive neuromonitors can alert the anesthesiologist and
monitoring techniques and advances in neuro- surgeon to allow modification of treatment strate-
logic, neuromuscular, and cardiovascular moni- gies. In OMS procedures, intricate work is often
toring further enhance the practitioner’s ability to done near major nerves and vascular structures,
provide safe and effective anesthesia. Topics which may require special anesthetic techniques.
selected for inclusion in this article focus on those Neuromonitoring techniques include cerebral
monitoring elements most germane to oral maxil- oxygen monitoring, processed electroencepha-
lofacial surgery (OMS). lography (pEEG), electromyography (EMG),
evoked potentials (EP), somatosensory EP
NEUROLOGIC MONITORING (SSEP), brainstem auditory EP, and visual EP.
Neural function is essential for cognition and to the This section explores the details of advances in
hallmark of human existence. A permanent loss neuromonitoring.
oralmaxsurgery.theclinics.com

Disclosure Statement: The authors have nothing to disclose.


a
Department of Anesthesiology and Critical Care Medicine, Baylor College of Medicine, 1 Baylor Plaza, MSC
120, Houston, TX 77030, USA; b Department of Anesthesiology, Baylor College of Medicine, 1 Baylor Plaza,
MSC 120, Houston, TX 77030, USA; c Department of Neurosurgery, Baylor College of Medicine, Houston,
TX, USA
* Corresponding author.
E-mail address: yd1@bcm.edu
twitter: @chrisdengMD (Y.D.)

Oral Maxillofacial Surg Clin N Am 31 (2019) 611–619


https://doi.org/10.1016/j.coms.2019.07.005
1042-3699/19/Ó 2019 Elsevier Inc. All rights reserved.
612 Deng et al

Cerebral Oxygenation Monitoring complex cardiac surgery to outpatient hernia


repair. Today, they are commonly used during
Cerebral oxygenation (O2) monitoring should be
general anesthesia cases to monitor the depth of
instituted during general anesthesia whenever
anesthesia. The most widely available device is
there is concern of intraoperative cerebral
the bispectral index monitor, which processes
ischemia, as in patients with known severe carotid
frontal electroencephalography signals to derive
stenosis, extremes in head positioning, and/or
a score between 0 and 100. Values greater than
existing intracranial abnormalities combined with
90 suggest wakefulness, whereas less than 60
large expected blood loss. There are many modal-
suggest general anesthesia.7
ities for directly monitoring cerebral O2 (ie, mixed
The pEEG monitors have been postulated to
jugular venous saturation, partial brain tissue O2),
ensure adequate depth of anesthesia, decrease
but these are invasive in nature and require a
the risk of intraoperative awareness, and postop-
neurosurgeon. One new technology marketed
erative cognitive dysfunction. So far, the evidence
over the last decade is near-infrared spectroscopy
is equivocal.8,9 Similar to NIRS, many factors
(NIRS), which places two small pads on the pa-
affect pEEG signal and therefore its interpretation.
tient’s forehead that continuously and noninva-
Nevertheless, if inhalational anesthetics are con-
sively monitor regional cerebral O2 saturation.1
tradicted because of either patient or surgical fac-
NIRS devices measure the transmission of near-
tors, pEEG monitoring may be helpful to guide the
infrared light and its differential absorption by
depth of anesthesia using intravenous (IV) anes-
chromophores (usually oxyhemoglobin and deox-
thetics during surgery.
yhemoglobin) across brain tissues. This absorp-
tion difference correlates to the degree of tissue
Electromyography
O2 saturation. Unlike conventional pulse oximetry,
NIRS interrogates arterial, venous, and capillary EMG is used to monitor cranial or peripheral
blood in totality as a weighted value that does nerves at risk of surgical injury, and is commonly
not require pulsatile flow to function. Currently it deployed during OMS procedures. EMG can iden-
is used to guide brain-protective strategies during tify direct mechanical trauma to the nerve, which is
cardiac surgery,2 carotid endarterectomy,3 endo- seen as high-frequency neurotonic discharges
vascular therapy for acute ischemic stroke,4 and (Fig. 1).10 In addition, it is used to identify nerves
traumatic brain injury monitoring.5 The normal that are otherwise difficult to visualize. When an
range of regional cerebral O2 saturation is between innervated muscle is triggered by stimulation, a
55% and 75%, although substantial intraindividual compound muscle action potential is recorded.11
and interindividual variability exists. A significant When monitoring EMG during spinal surgery, an
drop can suggest ongoing cerebral ischemia, increase in compound muscle action potential la-
which may necessitate maneuvers, such as raising tency can indicate nerve injury, and the surgeon
O2 delivery (eg, augmenting cardiac output [CO],
transfusing red blood cells) or lowering O2 extrac-
tion (eg, treating fever and infection, reducing work
of breathing).
Evidence supporting the routine use of NIRS is
currently limited. One must be wary that its value
can be influenced by many factors, such BP, he-
moglobin concentration, and cerebral blood vol-
ume. In addition, there is potential for “signal
contamination” by extracranial tissue (ie, scalp he-
matoma, scalp itself, or thick cranium).1,6 Further-
more, the sensitivity of global cerebral ischemia
detection is likely limited by the small sampling
area in the frontal cortex.1

Electroencephalography
Standard electroencephalography interpretation is
Fig. 1. (A) Electromyography recording showing a
cumbersome, requires professional interpretation, large isolated response from electric or mechanical
and is not routinely used in the operating room stimulation. (B) Nerve stretch or mechanical trauma
except in certain neurosurgical procedures. How- can cause continuous, high-frequency recordings
ever, in the last 20 years there has been an in- known as neurotonic discharges, suggestive of nerve
crease in pEEG monitoring, used anywhere from injury.
Advances in Anesthesia Monitoring 613

may need to act to mitigate further damage (ie, halt general, inhalational agents have greater effects
electrocautery activity in the proximity, decrease on EPs than do IV agents. Choice and dosage of
screw or plate tension). anesthetics should be tailored to the desired EP
Similarly, cranial nerve (CN) monitoring is a form monitoring technique. In addition, physiologic fac-
of EMG and shares the same principles, although tors (ie, anemia, hypothermia, hypotension, hyp-
only CNs with motor component can be monitored oxia) and patient positioning (ie, severe neck
(CN III, IV, V, VI, VII, IX, X, XI, and XII).12 flexion) can all affect EP assessment.

Evoked Potentials NEUROMUSCULAR MONITORING


EP monitoring is used to assess the integrity of a Neuromuscular blocking agents (NMBAs) or mus-
neural pathway. It typically involves stimulating pe- cle relaxants were introduced in 1942 by Griffith
ripheral sites of the body and recording responses and Johnson and are commonly used in OMS. In
centrally (opposite for motor EP [MEP]). The ampli- recent years, increased recognition of postopera-
tude and latency of such recorded waveforms are tive residual paralysis and its associated dangers
analyzed to provide functional neurologic assess- has led to greater emphasis on improved moni-
ment. A 50% decrease in waveform amplitude toring and routine reversal to decrease its inci-
and/or a 10% increase in latency may indicate dence. Newly published guidelines16 recommend
nerve dysfunction and/or injury, and necessitate the use of quantitative, objective measurements
modification in surgical strategy, patient posi- instead of traditional subjective measurements.
tioning, and anesthetic management.13 Subjective monitoring refers to visual or tactile
SSEP is the most commonly used EP monitoring evaluation of the train-of-four (TOF) count or
modality. An electrical stimulus is applied to a pe- degree of TOF fade in response to peripheral neu-
ripheral nerve in the upper or lower extremity and rostimulation (Fig. 3). It is the defacto standard
measured with scalp electrodes (Fig. 2). It is used today. Recent evidence indicates that a
particularly useful during spine surgery because TOF ratio greater than or equal to 0.9 (as opposed
it monitors the integrity of the dorsal root ganglia to the traditionally taught 0.7) is necessary for safe
and the dorsal columns.14 MEPs, however, use extubation and decreased postoperative residual
transcranial electrical stimulation and monitor the paralysis.17,18 However, physicians tend to over-
corticospinal tract, nerve roots, and peripheral estimate TOF ratio when using subjective evalua-
nerves.15 MEPs are more effective than SSEPs in tion19 and are unable to accurately detect fade
detecting motor neuron injury because changes when TOF ratio is greater than 0.4.20
there precede that of SSEPs. Other commonly Quantitative monitors include acceleromyogra-
used modalities include brainstem auditory EPs phy, which can accurately detect fade, TOF count,
for posterior fossa surgeries and visual EPs for and TOF ratio. It is a finger-sized device placed on
procedures near the occipital lobe. the thumb and measures its acceleration in
Most currently available anesthetics alter neural response to ulnar nerve stimulation. Some caveats
function by producing dose-dependent depres- to use include mandatory calibration and unim-
sion in synaptic activity. As such, the anesthetic ef- peded movement of the thumb during surgery.
fects vary with the location of the synapses. In Kinemyography is another quantitative monitor

Fig. 2. Typical neural pathways as-


sessed with SSEP and MEP moni-
toring. SSEPs are produced by
stimulation of a peripheral nerve
and measured centrally. MEPs are
produced by stimulation of the motor
cortex and measured peripherally.
NMJ, Neuromuscular junction.
614 Deng et al

Fig. 3. Neuromuscular blockade monitoring with TOF count and TOF fade. In profound block when TOF count is
0, one can provide tetanic stimulation for 5 seconds and count the number of posttetanic twitches to estimate
degree of blockade. Sugammadex can reverse deep blockade to a TOF count of 4 within 3 minutes, which is
not possible with neostigmine.

that measures the electrical signal generated from anesthetic management, such as preferred choice
the distortion of a mechanosensor, which is placed between succinylcholine and rocuronium for
between the base of the thumb and index finger. rapid-sequence induction when there are con-
Its results are not interchangeable with other quan- cerns for difficult airway management.
titative modalities, and the technology has similar
limitations as the acceleromyography.21 Finally, CARDIOVASCULAR MONITORING
previously described EMG can also objectively
monitor the neuromuscular function recovery, Until the last decade, cardiovascular monitoring in
although its use is much more cumbersome. anesthesia and surgery had essentially remained
Although there has not been a novel NMBA unchanged for many years. Conventional mea-
approved clinically in some time, sugammadex surement of heart rate and rhythm as depicted
has been introduced recently as a reversal by five-lead electrocardiogram and BP using
agent. Sugammadex is a g-cyclodextrin, which automated cuffs and/or intra-arterial catheter are
selectively binds free plasma steroidal NMBA well-accepted methods and have long been incor-
molecules, rendering them unavailable for redistri- porated into American Society of Anesthesiology
bution in the neuromuscular junction. Affinity is practice guidelines. For higher acuity patients,
greatest for rocuronium, followed by vecuronium, central venous access is required to either obtain
pancuronium, and pipecuronium.22 It offers rapid central venous pressure or allow for the insertion
and complete reversal (<3 minutes) in usual of a pulmonary artery catheter (PAC). More
clinical scenarios and can even reverse profound recently, however, there has been a substantial in-
rocuronium-induced blockade otherwise not crease in new technology centered around cardiac
possible with neostigmine (see Fig. 3).23 Currently monitoring, which has greatly expanded the clini-
it is not recommended in dialysis patients, and cian’s armamentarium and led to better under-
side effects although rare can include interference standing of patient management perioperatively.
with oral contraceptive pills, anaphylaxis, cardiac We briefly review these new methods categorized
arrhythmias,24 coronary vasospasm,25 and mild into minimally invasive and noninvasive groups,
prolongation of coagulation values.26 Neverthe- followed by a discussion on new markers of fluid
less, its availability has profound effects on responsiveness.
Advances in Anesthesia Monitoring 615

Minimally Invasive Cardiovascular Monitoring is useful in managing patients with severe lung dis-
ease. Pulse contour analysis continuously pro-
CO obtained from a PAC using thermodilution
vides real-time CO monitoring, although external
calculations has been considered gold standard
recalibration is required periodically using tradi-
for the last 40 years.27 However, because of con-
tional thermodilution methods. Because central
cerns about its invasive nature, perceived deficits
venous access is necessary for its function, it
in efficacy, and applicability, PAC use has gener-
has since given way to less invasiveness tech-
ally been declining since the 1990s.28,29 In the
niques, such as LiDCO and FloTrac.
mid-2000s, several devices have emerged
LiDCOplus system (LiDCO, London, UK) is
including PiCCO (Pulsion Medical System, Feld-
another calibrated device similar to PiCCO. How-
kirchen, Germany), LiDCO (LiDCO, London, UK),
ever, instead of using cold saline for thermodilution
and FloTrac/Vigileo (Edwards Lifesciences Irvine,
CO calculation, it uses lithium as an indicator.35
CA, USA), which use the pulse contour analysis
This allows it to function without a central line,
technology and made it feasible to derive stroke
because lithium can be injected through a periph-
volume (SV) and CO monitoring without the risks
eral IV and picked up by a sensor attached to the
of PAC insertion.30,31
arterial line. The device is recalibrated every
Pulse contour analysis uses waveforms
8 hours (similar to PiCCO). Disadvantages include
captured by the arterial line transducer and feeds
chronic lithium therapy and nondepolarizing
them into a proprietary algorithm unique to each
NMBAs invalidate results, and lithium is contrain-
device. The output variables include SV, SV varia-
dicated in first trimester pregnancy.36–38 In gen-
tion, CO, and systemic vascular resistance. The
eral, LiDCO is also well validated in a variety of
first device to market using this technology was
patient populations, and is precise compared
the PiCCO system in 2000. PiCCO system still
with the other systems described here.35,39,40
required a central line and arterial line to function,
Finally, one of the newest devices to market is
but was generally well validated across diverse pa-
the FloTrac/Vigileo system. It is unique in that it
tient populations and useful in certain groups,
is an uncalibrated system, and calculates CO
such as children where PAC is too large to be
and SV using pulse contour analysis alone. To do
inserted.32–34 Another advantage of the PiCCO
so it factors in patient’s age, gender, weight, and
system is that intrathoracic blood volume and
height, and inputs these variables along with the
extravascular lung water can be calculated, which

Table 1
Minimally invasive and noninvasive cardiac output monitoring devices

Device Name Technology Used Advantage Disadvantage


Minimally Invasive
PiCCO Pulse contour Continuous CO monitoring, Central access required
analysis external calibration for
precision, well validated
LiDCO Pulse contour Continuous CO monitoring, Lithium injection required,
analysis external calibration for contraindicated in pregnancy,
precision, well validated, can interfere with chronic
no central access lithium therapy or NMBA
FloTrac/ Pulse contour Continuous CO monitoring, Least accurate of the 3,
Vigileo analysis easy to use, no calibration inconsistent tracking, prone
required to drift over time
Noninvasive
ClearSight Pulse contour Finger cuff used without need Not well validated
analysis for intra-arterial access
TEB Bioimpedance Continuous CO monitoring, Not well validated, changes in
easy to place thoracic fluid volume can
impede with results
TTE Ultrasound, Highly accurate, well validated, Operator dependent, requires
Doppler easy to establish diagnosis in training, only intermittent
experienced hands measurement

Abbreviations: TEB, thoracic electrical bioimpedance; TTE, transthoracic echocardiography.


616 Deng et al

patient’s hemodynamic profile into its software al- transducer setup), it has gained extensive popu-
gorithm against a dataset of internally validated larity across a wide variety of operating room
subjects. Because it lacks external calibration, it and intensive care unit cases (Table 1).
is prone to drift and has limited accuracy in criti-
cally ill patients with wide swings in systemic
Noninvasive Cardiovascular Monitoring
vascular resistance, severe arrhythmias, aortic
regurgitation, and poor arterial waveform.41 The Two noninvasive CO monitoring techniques devel-
company has so far released four iterations of its oped recently include the ClearSight system
algorithm software and has improved its accuracy (Edwards Lifesciences) and thoracic electrical bio-
in hyperdynamic and vasoplegic patients, but not impedance. ClearSight uses the same pulse con-
in cardiac, liver transplantation, or abdominal tour analysis principle but uses finger cuffs with
aortic aneurysm surgeries.42–45 Despite these con- LED emitters to obtain waveforms instead of
cerns, because of its ease of implementation (Flo- intra-arterial catheters. Thoracic electrical bio-
Trac adapter can be connected to existing arterial impedance places electrodes across the thorax

Fig. 4. The basic 4 views of Focus Assessed Transthoracic Echocardiography (FATE) protocol. AO, aorta; LA, left
atrium; LV, left ventricle; RA, right atrium; RV, right ventricle. (From Oveland N, Bogale N, Waldron B, et al. Focus
assessed transthoracic echocardiography (FATE) to diagnose pleural effusions causing haemodynamic compro-
mise. Case Reports in Clin Med. 2013;2(3):190.)
Advances in Anesthesia Monitoring 617

and detects changes in electrical impedance as Table 2


blood flow in the thoracic aorta varies during Dynamic indices of fluid responsiveness and
cardiac cycle. The limitations of both devices their accuracy
include narrow patient selection, high percent er-
ror compared with PACs, and low overall Sensitivity Specificity
precision.46–48 (%) (%) AUC
In recent years, point-of-care ultrasound has PPV >12% 63 92 0.81
surged in popularity because of the greater avail-
SPV >9% 47 92 0.82
ability and declining cost of portable ultrasound
SVV >10% 56 69 0.70
machines. Perioperative transthoracic echocardi-
ography (TTE) brings another dimension to car- IVCci >15% 31 97 0.62
diovascular monitoring, enabling a clinician to DSV >12% 69 89 0.90
more accurately assess volume status, cardiac
Abbreviations: DSV, change in stroke volume after fluid
function, and presence of structural heart dis- challenge; AUC, area under the receiver operating charac-
ease. A complete echocardiographic examination teristic curve; IVCci, inferior vena cava collapsibility index;
requires significant training and expertise. How- SPV, systolic pressure variation; SVV, stroke volume
ever, multiple abridged protocols have been variation.
developed, such the Focus Assessed Transtho-
racic Echocardiography and Rapid Assessment
by Cardiac Echo, to allow perioperative clinicians collapsibility index, E wave velocity, and aortic ve-
rapid evaluation of key identifiers of cardiovascu- locity time index variations.56 Each of these
lar aberrations.49,50 In Focus Assessed Transtho- markers has independent cutoff values for sensi-
racic Echocardiography protocol, for example, tivity and specificity (Table 2). For example, a hy-
only four views are necessary for a complete eval- potensive patient with concomitant PPV greater
uation (Fig. 4). TTE is also superior to catheter- than 12% suggests that the patient can benefit
based pressure measurements (eg, PAC) in that from fluid resuscitation. One must be aware of
cardiac chamber volume can now be directly the pitfalls of each marker in specific clinical sce-
calculated instead of inferred from pressure- narios, such as decreased accuracy of PPV in
volume curve. Unlike the pulse contour analysis spontaneously breathing patients or those with
technique, TTE can directly measure SV and CO high intrathoracic or intra-abdominal pressures.
and is much less prone to biases. Lastly, point- Nevertheless, these dynamic markers of fluid
of-care ultrasound can make rapid and accurate resuscitation have proven superiority over static
diagnosis of other systemic pathologies, such as measures, and have shown a high correlation
pneumothorax, consolidations, effusions, and with PAC and TTE.56 When used with goal-
intra-abdominal hemorrhage, thus making it directed fluid therapy, it could potentially reduce
an invaluable tool in the modern perioperative the amount of fluid resuscitation, decrease length
setting. of stay, postoperative ileus, and prolonged me-
chanical ventilation, resulting in enhanced recov-
ery after surgery and anesthesia.55,57
DYNAMIC MARKERS OF RESUSCITATION
Given the penetration of these cardiovascular SUMMARY
monitoring devices, multiple new markers of dy-
namic volume responsiveness have since come In this article, we provide a concise and focused
into vogue. It has been recognized that 40% of review on the new advances in perioperative
critically ill and hypotensive patients are actually neurologic, neuromuscular, and cardiovascular
fluid responsive and can benefit from fluid ther- monitoring. It is imperative for surgeons and anes-
apy, whereas excess fluid loading can increase thesiologists to have detailed understanding of the
mortality.51,52 Traditional static measurements of function and limitation of each device to safely
fluid status, such as central venous pressure, guide patients through their surgeries.
have been shown to be extremely poor predictors
of volume status and volume responsiveness.53,54 REFERENCES
The most commonly used dynamic markers of
fluid resuscitation include pulse pressure variation 1. Ghosh A, Elwell C, Smith M. Review article: cerebral
(PPV), SV variation, and systolic pressure varia- near-infrared spectroscopy in adults: a work in prog-
tion, all of which are obtained with the new ress. Anesth Analg 2012;115(6):1373–83.
pulse contour analysis devices.55 Similarly, TTE 2. Zheng F, Sheinberg R, Yee M-S, et al. Cerebral near-
can provide clinicians with inferior vena cava infrared spectroscopy monitoring and neurologic
618 Deng et al

outcomes in adult cardiac surgery patients: a sys- the train of four ratio of the adductor pollicis muscle
tematic review. Anesth Analg 2013;116(3):663–76. from neuromuscular blockade. Anesth Analg 2006;
3. Badenes R, Garcı́a-Pérez ML, Bilotta F. Intraopera- 102(3):937–42.
tive monitoring of cerebral oximetry and depth of 18. Eikermann M, Groeben H, Hüsing J, et al. Acceler-
anaesthesia during neuroanesthesia procedures. ometry of adductor pollicis muscle predicts recovery
Curr Opin Anaesthesiol 2016;29(5):576–81. of respiratory function from neuromuscular blockade.
4. Goyal M, Demchuk AM, Menon BK, et al. Random- Anesthesiology 2003;98(6):1333–7.
ized assessment of rapid endovascular treatment 19. Brull SJ, Silverman DG. Visual and tactile assess-
of ischemic stroke. N Engl J Med 2015;372(11): ment of neuromuscular fade. Anesth Analg 1993;
1019–30. 77(2):352–5.
5. Leal-Noval SR, Cayuela A, Arellano-Orden V, et al. 20. Capron F, Fortier L-P, Racine S, et al. Tactile fade
Invasive and noninvasive assessment of cerebral detection with hand or wrist stimulation using train-
oxygenation in patients with severe traumatic brain of-four, double-burst stimulation, 50-hertz tetanus,
injury. Intensive Care Med 2010;36(8):1309–17. 100-hertz tetanus, and acceleromyography. Anesth
6. Davie SN, Grocott HP. Impact of extracranial Analg 2006;102(5):1578–84.
contamination on regional cerebral oxygen satura- 21. Stewart PA, Freelander N, Liang S, et al. Comparison
tion: a comparison of three cerebral oximetry tech- of electromyography and kinemyography during
nologies. Anesthesiology 2012;116(4):834–40. recovery from non-depolarising neuromuscular
7. Liu J, Singh H, White PF. Electroencephalographic blockade. Anaesth Intensive Care 2014;42(3):
bispectral index correlates with intraoperative recall 378–84.
and depth of propofol-induced sedation. Anesth An- 22. Bom A, Hope F, Rutherford S, et al. Preclinical phar-
alg 1997;84(1):185–9. macology of sugammadex. J Crit Care 2009;24(1):
8. Punjasawadwong Y, Phongchiewboon A, Bunchung 29–35.
mongkol N. Bispectral index for improving anaes- 23. Sørensen MK, Bretlau C, Gätke MR, et al. Rapid
thetic delivery and postoperative recovery. Co- sequence induction and intubation with rocuronium-
chrane Database Syst Rev 2014;(6):CD003843. sugammadex compared with succinylcholine: a ran-
9. Messina AG, Wang M, Ward MJ, et al. Anaesthetic domized trial. Br J Anaesth 2012;108(4):682–9.
interventions for prevention of awareness during sur- 24. Bhavani SS. Severe bradycardia and asystole after
gery. Cochrane Database Syst Rev 2016;(10): sugammadex. Br J Anaesth 2018;121(1):95–6.
CD007272. 25. Okuno A, Matsuki Y, Tabata M, et al. A suspected
10. Holland NR. Intraoperative electromyography. J Clin case of coronary vasospasm induced by anaphy-
Neurophysiol 2002;19(5):444–53. lactic shock caused by rocuronium-sugammadex
11. López JR. Neurophysiologic intraoperative moni- complex. J Clin Anesth 2018;48:7.
toring of the oculomotor, trochlear, and abducens 26. Rahe-Meyer N, Fennema H, Schulman S, et al. Ef-
nerves. J Clin Neurophysiol 2011;28(6):543–50. fect of reversal of neuromuscular blockade with su-
12. Jameson LC, Sloan TB. Monitoring of the brain and gammadex versus usual care on bleeding risk in a
spinal cord. Anesthesiol Clin 2006;24(4):777–91. randomized study of surgical patients. Anesthesi-
13. Toleikis JR, American Society of Neurophysiological ology 2014;121(5):969–77.
Monitoring. Intraoperative monitoring using somato- 27. Swan HJ, Ganz W, Forrester J, et al. Catheterization
sensory evoked potentials. A position statement by of the heart in man with use of a flow-directed
the American Society of Neurophysiological Moni- balloon-tipped catheter. N Engl J Med 1970;
toring. J Clin Monit Comput 2005;19(3):241–58. 283(9):447–51.
14. Banoub M, Tetzlaff JE, Schubert A. Pharmacologic 28. Gore JM, Goldberg RJ, Spodick DH, et al.
and physiologic influences affecting sensory evoked A community-wide assessment of the use of pulmo-
potentials: implications for perioperative monitoring. nary artery catheters in patients with acute myocar-
Anesthesiology 2003;99(3):716–37. dial infarction. Chest 1987;92(4):721–7.
15. Lall RR, Lall RR, Hauptman JS, et al. Intraoperative 29. Connors AF, Speroff T, Dawson NV, et al. The effec-
neurophysiological monitoring in spine surgery: indi- tiveness of right heart catheterization in the initial
cations, efficacy, and role of the preoperative check- care of critically ill patients. SUPPORT Investigators.
list. Neurosurg Focus 2012;33(5):E10. JAMA 1996;276(11):889–97.
16. Checketts MR, Alladi R, Ferguson K, et al. Recom- 30. Sangkum L, Liu GL, Yu L, et al. Minimally invasive or
mendations for standards of monitoring during noninvasive cardiac output measurement: an up-
anaesthesia and recovery 2015: Association of date. J Anesth 2016;30(3):461–80.
Anaesthetists of Great Britain and Ireland. Anaes- 31. Pugsley J, Lerner AB. Cardiac output monitoring: is
thesia 2016;71(1):85–93. there a gold standard and how do the newer tech-
17. Eikermann M, Blobner M, Groeben H, et al. Postop- nologies compare? Semin Cardiothorac Vasc
erative upper airway obstruction after recovery of Anesth 2010;14(4):274–82.
Advances in Anesthesia Monitoring 619

32. Goedje O, Hoeke K, Lichtwarck-Aschoff M, et al. abdominal aortic aneurysm surgery. J Cardiothorac
Continuous cardiac output by femoral arterial ther- Vasc Anesth 2012;26(2):223–6.
modilution calibrated pulse contour analysis: com- 45. Biancofiore G, Critchley LAH, Lee A, et al. Evaluation
parison with pulmonary arterial thermodilution. Crit of an uncalibrated arterial pulse contour cardiac
Care Med 1999;27(11):2407–12. output monitoring system in cirrhotic patients under-
33. Tibby SM, Hatherill M, Marsh MJ, et al. Clinical vali- going liver surgery. Br J Anaesth 2009;102(1):47–54.
dation of cardiac output measurements using 46. Bogert LWJ, Wesseling KH, Schraa O, et al. Pulse
femoral artery thermodilution with direct Fick in contour cardiac output derived from non-invasive
ventilated children and infants. Intensive Care Med arterial pressure in cardiovascular disease. Anaes-
1997;23(9):987–91. thesia 2010;65(11):1119–25.
34. Buhre W, Weyland A, Kazmaier S, et al. Comparison 47. Stover JF, Stocker R, Lenherr R, et al. Noninvasive
of cardiac output assessed by pulse-contour anal- cardiac output and blood pressure monitoring
ysis and thermodilution in patients undergoing mini- cannot replace an invasive monitoring system in crit-
mally invasive direct coronary artery bypass ically ill patients. BMC Anesthesiol 2009;9:6.
grafting. J Cardiothorac Vasc Anesth 1999;13(4): 48. Porhomayon J, El-Solh A, Papadakos P, et al. Car-
437–40. diac output monitoring devices: an analytic review.
35. Linton R, Band D, O’Brien T, et al. Lithium dilution car- Intern Emerg Med 2012;7(2):163–71.
diac output measurement: a comparison with ther- 49. McLean A, Yastrebov K. Echocardiography training
modilution. Crit Care Med 1997;25(11):1796–800. for the intensivist. Crit Care Resusc 2007;9(4):
36. Reuter DA, Huang C, Edrich T, et al. Cardiac output 319–22.
monitoring using indicator-dilution techniques: ba- 50. Jensen MB, Sloth E, Larsen KM, et al. Transthoracic
sics, limits, and perspectives. Anesth Analg 2010; echocardiography for cardiopulmonary monitoring
110(3):799–811. in intensive care. Eur J Anaesthesiol 2004;21(9):
700–7.
37. Ostergaard D, Engbaek J, Viby-Mogensen J.
51. Boyd JH, Forbes J, Nakada T, et al. Fluid resuscita-
Adverse reactions and interactions of the neuromus-
tion in septic shock: a positive fluid balance and
cular blocking drugs. Med Toxicol Adverse Drug
elevated central venous pressure are associated
Exp 1989;4(5):351–68.
with increased mortality. Crit Care Med 2011;39(2):
38. Pearse RM, Ikram K, Barry J. Equipment review: an
259–65.
appraisal of the LiDCO plus method of measuring
52. Kanji HD, McCallum J, Sirounis D, et al. Limited
cardiac output. Crit Care 2004;8(3):190–5.
echocardiography-guided therapy in subacute
39. Costa MG, Della Rocca G, Chiarandini P, et al.
shock is associated with change in management
Continuous and intermittent cardiac output mea-
and improved outcomes. J Crit Care 2014;29(5):
surement in hyperdynamic conditions: pulmonary
700–5.
artery catheter vs. lithium dilution technique. Inten-
53. Kumar A, Anel R, Bunnell E, et al. Pulmonary artery
sive Care Med 2008;34(2):257–63.
occlusion pressure and central venous pressure fail
40. Hadian M, Kim HK, Severyn DA, et al. Cross-com- to predict ventricular filling volume, cardiac perfor-
parison of cardiac output trending accuracy of mance, or the response to volume infusion in normal
LiDCO, PiCCO, FloTrac and pulmonary artery cathe- subjects. Crit Care Med 2004;32(3):691–9.
ters. Crit Care 2010;14(6):R212. 54. Marik PE, Monnet X, Teboul J-L. Hemodynamic pa-
41. Mayer J, Boldt J, Poland R, et al. Continuous arterial rameters to guide fluid therapy. Ann Intensive Care
pressure waveform-based cardiac output using 2011;1(1):1.
the FloTrac/Vigileo: a review and meta-analysis. 55. Bednarczyk JM, Fridfinnson JA, Kumar A, et al.
J Cardiothorac Vasc Anesth 2009;23(3):401–6. Incorporating dynamic assessment of fluid respon-
42. De Backer D, Ospina-Tascon G, Salgado D, et al. siveness into goal-directed therapy: a systematic re-
Monitoring the microcirculation in the critically ill pa- view and meta-analysis. Crit Care Med 2017;45(9):
tient: current methods and future approaches. Inten- 1538–45.
sive Care Med 2010;36(11):1813–25. 56. Chaves RCF, Corrêa TD, Neto AS, et al. Assessment
43. Desebbe O, Henaine R, Keller G, et al. Ability of the of fluid responsiveness in spontaneously breathing
third-generation FloTrac/Vigileo software to track patients: a systematic review of literature. Ann Inten-
changes in cardiac output in cardiac surgery pa- sive Care 2018;8(1):21.
tients: a polar plot approach. J Cardiothorac Vasc 57. Berger MM, Gradwohl-Matis I, Brunauer A, et al. Tar-
Anesth 2013;27(6):1122–7. gets of perioperative fluid therapy and their effects
44. Kusaka Y, Yoshitani K, Irie T, et al. Clinical compari- on postoperative outcome: a systematic review
son of an echocardiograph-derived versus pulse and meta-analysis. Minerva Anestesiol 2015;81(7):
counter-derived cardiac output measurement in 794–808.

You might also like