bloqueadores neuromusculares actualmente

Embed Size (px)

Citation preview

  • 1332 www.ccmjournal.org May 2013 Volume 41 Number 5

    Intensivists use neuromuscular blocking agents for a variety of clin-ical conditions, including for emergency intubation, acute respira-tory distress syndrome, status asthmaticus, elevated intracranial pressure, elevated intra-abdominal pressure, and therapeutic hypothermia after ventricular fibrillationassociated cardiac arrest. The continued creation and use of evidence-based guidelines and protocols could ensure that neuromuscular blocking agents are used and monitored appropriately. A collaborative multidisciplinary approach coupled with constant review of the pharmacology, dos-ing, drug interactions, and monitoring techniques may reduce the adverse events associated with the use of neuromuscular block-ing agents. (Crit Care Med 2013; 41:13321344)Key Words: associated complications; ICU; monitoring; neuromuscular blockade; pharmacology; reversal; use

    Currently, controversy surrounds the use of neuromus-cular blocking agents (NMBAs) in the ICU. Proposed adjunctive applications for NMBAs in the ICU include the following: improving chest wall compliance, eliminating ventilator dyssynchrony, reducing intra-abdominal pressures (IAPs), preventing and treating shivering, and preventing ele-vations in intracranial pressure (ICP) from airway stimulation. These relative indications are tempered by clinician concern for development of prolonged ICU-acquired weakness (ICU-AW); prolonged mechanical ventilation; the risk of patient awareness during paralysis; and an increased risk for deep venous throm-bosis (DVT), corneal abrasions, and anaphylaxis (1).

    Today, NMBAs are being used much less frequently than in past years. A survey of 34 ICUs from the 1980s reported that 90% of mechanically ventilated patients received NMBAs

    (2). A more recent international observational study reported that only 13% of patients on mechanical ventilation received NMBAs (3). A 2006 Canadian survey of critical care practitio-ner practices noted that the most important factors for choos-ing a specific NMBA were clinician experience, duration of action, mechanism of action, and patient-specific factors (4). A similar survey conducted in the United States reported that clinical experience and perception were the most common rea-sons for choosing a NMBA (5).

    This review examines the anatomy of the NMJ and the physiology of neuromuscular transmission. The classification, pharmacology, reversal, monitoring, associated complications, and guidelines concerning NMBAs will also be reviewed. In addition, the use of NMBAs in critically ill patients during six clinical situations will be examined: emergency intubation, acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), status asthmaticus, elevated ICP, elevated IAP, and therapeutic hypothermia after out-of-hospital ventricular fibrillation (VF)-associated cardiac arrest (1, 68). A com-puterized literature search through MEDLINE and PubMed (January 1975September 2012) was performed to identify observational and interventional studies evaluating NMBAs for management of emergent/urgent intubation, ALI/ARDS, status asthmaticus, elevated ICP, elevated IAP, and therapeutic hypothermia. The following search terms were used: NMBAs, neuromuscular blockers, paralytics, paralysis, succinylcho-line, atracurium, cisatracurium, pancuronium, rocuronium, vecuronium, critical care, intensive care, mechanical ventila-tion, ALI, ARDS, status asthmaticus, ICP, traumatic brain injury, IAP, abdominal compartment syndrome (ACS), thera-peutic hypothermia, train-of-four (TOF), peripheral nerve stimulator, monitoring, complications, DVT, corneal abrasions, anaphylaxis and neuromuscular blockade, and reversal. We excluded articles published in languages other than English.

    ANATOMY/PHYSIOLOGY OF NEUROMUSCULAR TRANSMISSIONThere are three major components involved in neuromuscular transmission: the neuron, the neurotransmitter (acetylcho-line), and the muscle fiber (9). Figure 1 depicts the relation-ship between these three major components of the NMJ. The NMJ marks the location where the axonal terminus lies in close

    The Use of Neuromuscular Blocking Agents in the ICU: Where Are We Now?

    Steven B. Greenberg, MD1,2; Jeffery Vender, MD, MBA, FCCP, FCCM1,2

    1University of Chicago, Chicago, IL.2NorthShore University HealthSystem, Evanston, IL.

    Dr. Vendor consulted for Pharmadiem. Dr. Greenberg has disclosed that he does not have any potential conflicts of interest.

    For information regarding this article, E-mail: [email protected]

    Critical Care Medicine

    0090-3493

    10.1097/CCM.0b013e31828ce07c

    41

    5

    00

    00

    2013

    Copyright 2013 by the Society of Critical Care Medicine and Lippincott Williams & Wilkins

    DOI: 10.1097/CCM.0b013e31828ce07c

    Esther

    Concise Definitive ReviewSeries Editor, Jonathan E. Sevransky, MD, MHS

  • Concise Definitive Review

    Critical Care Medicine www.ccmjournal.org 1333

    proximity to the muscle membrane. The motor unit comprises a lower motor neuron and its associated innervated muscle. One nerve has the capability of innervating over 100 muscle fibers in the motor unit. Acetylcholine, a neurotransmitter, is produced and stored in vesicles at the NMJ and then released into the synapse once an action potential is generated (9, 10).

    Once the action potential reaches the motor terminus, the depolarization results in the release (in the presence of calcium) of approximately 4 million molecules of acetylcholine into the NMJ. Two acetylcholine molecules bind to a postsynaptic nico-tinic receptor on the muscle resulting in an excitatory postsyn-aptic end-plate potential. Once this endplate potential reaches its threshold, it produces a muscle action potential that results in muscle contraction. The muscle returns to its precontractile state once acetylcholine is broken down by the enzyme, ace-tylcholinesterase. The large amount of acetylcholine molecules released in the NMJ acts as a protective mechanism to ensure muscle contraction (911).

    CLASSIFICATION OF NMBAs AND PHARMACODYNAMICSMuscle relaxants are typically classified by their mechanism of action (depolarizing vs nondepolarizing) and duration of action (short, intermediate, and long acting). Table 1 depicts the pharmacokinetics, pharmacodynamics, and data relating to dosing and mode of administration of commonly used NMBAs in the ICU (1114). Succinylcholine, the only avail-able depolarizing agent, binds to acetylcholine receptors and causes persistent depolarization (10). Succinylcholine pro-duces initial muscle fasciculations followed by flaccid paralysis (10). It is frequently used for urgent or emergent intuba-tion in the ICU. Nondepolarizing agents act as competitive antagonists of nicotinic receptors, blocking the action of ace-tylcholine (11). These nondepolarizing drugs can be further divided into two groups, aminosteroids (i.e., pancuronium,

    vecuronium, and rocuronium) and benzylisoquinoliniums (i.e., atracurium, cisatracurium, doxacurium, and mivacu-rium). Each drug within these classifications has unique phar-macokinetic properties (1113).

    PharmacologyAltered Physiology During Critical Illness. Critical illness is associated with altered physiology that may affect the pharma-cokinetics of NMBAs. A lack of understanding or recognition of these changes can result in clinical failures or adverse drug events (15). This section will discuss how age, hypothermia, and electrolytes/pH disturbances may alter the action of NMBAs.

    Multiple factors may affect the pharmacokinetics of NMBAs in critically ill patients. Age-related changes (i.e., reduction in total body water, lean body mass, and serum albumin concen-tration) may result in a reduction in the volume of distribution of NMBAs (13). Increased age may contribute to a reduction in the rate of drug elimination via decreases in cardiac output, glo-merular filtration rate, and liver function (1215). Hypothermia may prolong the duration of action of nondepolarizing NMBAs (16, 17). The mechanisms underlying this prolongation include an altered sensitivity of the NMJ, a reduction in acetylcholine mobilization, decreased muscle contractility, reduced renal and hepatic excretion, changes in drug volumes of distributions, and pH-related changes at the NMJ (1618). Hypothermia may also slow the Hoffmann elimination process and prolong the effect of both cisatracurium and atracurium (1318).

    Electrolyte and pH disturbances may alter the duration of action of NMBAs. Hypokalemia may augment the block-ade induced by nondepolarizing agents, while also reduc-ing the ability of neostigmine (and other anticholinesterase reversal agents) to reverse the blockade (12, 13). Magnesium prolongs the duration of neuromuscular blockade by inhibit-ing calcium channels in the presynaptic terminal and inhibit-ing postjunctional potentials. Calcium triggers acetylcholine

    Figure 1. Depicts the major components of the neuromuscular junction: the neuron, acetylcholine, and the muscle. Acetylcholine is stored in vesicles within the neuron. When an action potential is generated, acetylcholine is released into the synapse in a process called exocytosis. Acetylcholine binds to receptors on the muscle cell to stimulate muscle contraction. (Reproduced with permission from Bargmann C: Neurosciences: Genomics reaches the synapse. Nature 2005; 436:473474.)

  • Greenberg and Vender

    1334 www.ccmjournal.org May 2013 Volume 41 Number 5

    release, and therefore, hypercalcemia may both reduce the sen-sitivity to NMBA and decrease the duration of neuromuscular blockade. The blockade effect of nondepolarizing agents may be enhanced (although the mechanism is not clearly under-stood) by both respiratory and metabolic acidosis (12, 13). Antagonism of neuromuscular blockade by reversal agents may occur more slowly in the presence of a respiratory acidosis or metabolic alkalosis (12, 13).

    Organ Dysfunction and NMBAs. Aminosteroid NMBA pharmacokinetics can be affected by liver dysfunction. Pan-curonium half-life is prolonged in patients with cirrhosis because of its increased volume of distribution (19). Similarly, vecuronium can also have a prolonged elimination half-life, reduced clearance, and enhanced blockade in the setting of cir-rhosis. However, the elimination and clearance of vecuronium may not be altered in less severe forms of liver disease (13). Severe hepatic dysfunction can also increase the volume of dis-tribution of rocuronium, which will prolong its elimination half-life and recovery time after reversal (13). Large intubating

    doses (1.2 mg/kg) or repeated doses of rocuronium in the set-ting of severe liver disease typically prolong its effect (13).

    Aminosteroid NMBAs have variable durations of action in the setting of severe renal disease (13, 19). Renal failure increases elimination half-life and decreases clearance most significantly for the long-acting aminosteroid muscle relaxants (i.e., pancuronium) (13). Vecuronium may have a prolonged effect due to its active metabolite, 3-desacetyl vecuronium, which accumulates to a significant degree with severe renal dis-ease (19). Rocuronium can have an increased volume of distri-bution and decreased plasma clearance in the setting of renal failure (21, 22). However, the duration of action of repeated or single doses of rocuronium is typically not affected by renal failure (13, 21, 22).

    Benzylisoquinoliniums may be preferable in ICU patients as they are not affected by renal or hepatic disease (13, 19). Hoffman degradation (a nonenzymatic degradation) accounts for most of the metabolism of both atracurium and cisatracurium (19). Cisatracurium ICU infusion studies (> 24-hr duration) suggest

    TABLE 1. Drug Information on the Commonly Used Neuromuscular Blocking Agents in the ICU

    NMBA Agent

    NMBA Type

    Pancuronium

    A

    Vecuronium

    A

    Rocuronium

    A

    Atracurium

    B

    Cisatracurium

    B

    Succinylcholine

    D

    Category (acting)

    Long acting Intermediate Intermediate Intermediate Intermediate Short

    Time to maximal blockade (min)

    23 34 12 35 23 < 1

    Duration of action (min)

    60100 2035 2035, 6080 with rapid sequence dose

    2035 3060 510

    Dose

    Bolus 0.050.1 mg/kg 0.080.1 mg/kg 0.61 mg/kg (11.2 mg/ kg for rapid sequence)

    0.40.5 mg/kg 0.l0.2 mg/kg 1 mg/kg, dose higher in pediatrics

    Continuous infusion dosing

    0.81.7 mcg/ kg/min

    0.81.7 mcg/ kg/min

    812 mcg/ kg/min

    520 mcg/ kg/min

    13 mcg/kg/ min

    Infusions no longer used commonly

    Elimination 4570% renal, 1015% hepatic

    50% renal, 3550% hepatic

    33% renal, < 75% hepatic

    510% renal, Hoffman elimination

    510% renal, Hoffman elimination

    Plasma cholinesterase

    Active metabolites

    3-OH and 17-OH pancuronium

    3-Desacetyl- Vecuronium

    None None (toxic metabolite- laudanosine)

    None None

    Side effects Vagal blockade, sympathetic stimulation, blocks muscarinic stimulation (bradycardia)

    Vagal blockade at higher doses

    Vagal blockade at higher doses, weakly blocks muscarinic stimulation (bradycardia)

    Histamine release, minimal ganglionic blockade

    None Minimal amount of histamine release, muscarinic stimulation (bradycardia)

    NMBA = neuromuscular blocking agent, A = aminosteroid, B = benzylisoquinolinium, D = depolarizing agent.Information taken from references 1526.

  • Concise Definitive Review

    Critical Care Medicine www.ccmjournal.org 1335

    that there is marginal accumulation of the drug. Several stud-ies suggest that the duration of action of cisatracurium is not affected by organ dysfunction (23). Laudanosine, a metabolite of atracurium, has central nervous system stimulating effects. Laudanosine levels have been reported to increase when using long-term atracurium infusions but are typically not high enough to promote seizure activity in humans (13, 19, 23).

    Drug Interactions and NMBAs. Drug interactions may also alter the duration of action of NMBAs. Table 2 depicts some of the more common effects of drugs on neuromus-cular blockade. Certain antibiotics (i.e., aminoglycosides, clindamycin, and tetracycline) prolong the duration of NMBAs (24). Furosemide, calcium channel blockers, local anesthetics, and inhaled anesthetics can also potentiate neuromuscular blockade. Patients on chronic antiepileptic medications are typically resistant to aminosteroid NMBAs. This phenomenon may be due to increase clearance of these

    NMBAs, an increase in binding to -1 acid glycoproteins, or an up-regulation of acetylcholine receptors. Participa-tion by a clinical pharmacist in the multidisciplinary ICU daily rounds coupled with the use of close neuromuscular monitoring may help prevent unwanted prolongation or reduction in neuromuscular blockade produced by drug interactions (1, 13, 24).

    Reversal AgentsAnticholinesterase reversal agents (neostigmine, edropho-nium, or pyridostigmine) are only effective for nondepolariz-ing blockers and increase acetylcholine at the NMJ to reduce residual neuromuscular blockade (25). The duration of ace-tylcholinesterase inhibition is longer with pyridostigmine and neostigmine when compared with edrophonium, due to a strong covalent bond interaction between these two drugs and acetylcholinesterase (26, 27).

    TABLE 2. Drug Interactions and Neuromuscular Blocking Agents

    Drug Method

    Causes resistance to NMB

    Phenytoin Up-regulation of acetylcholine-R

    Ranitidine Unknown

    Carbamazepine Competes for acetylcholine receptors

    Theophylline, caffeine Unknown

    Calcium Antagonizes potentiating effect of magnesium on neuromuscular blockade

    Prolongs NMB

    Inhaled anesthetics Reduced postjunctional receptor sensitivity to acetylcholine

    Magnesium Competes with calcium presynaptically

    Potassium Low potassium enhances blockade and reduces ability of reversal by antagonists

    Lithium Activation of potassium channels presynaptically

    Immunosuppressant

    Corticosteroids May decrease sensitivity of endplate to acetylcholine

    Cyclosporine May inhibit neuromuscular blocking agent metabolism

    Cardiovascular Reduces prejunctional acetylcholine release and decreases muscle contractility

    Furosemide

    -blockers and calcium channel blockers

    Procainamide

    Quinidine

    Antibiotics Reduces prejunctional acetylcholine release, decreases postjunctional receptor sensitivity to acetylcholine, blocks acetylcholine receptors, or disrupts ion channels

    Aminoglycosides

    Tetracyclines

    Clindamycin

    Vancomycin

    NMB = neuromuscular blocker.Information produced from references (15, 16, 23, 24).

  • Greenberg and Vender

    1336 www.ccmjournal.org May 2013 Volume 41 Number 5

    Concomitant use of antimuscarinics (glycopyrrolate or atropine) with anticholinesterases is recommended to coun-teract side effects of increasing acetylcholine at muscarinic sites (i.e., bradycardia, bradyarrhythmias, increased secretions, and bronchoconstriction) (26, 27). Atropine (0.0050.01 mg/kg) is typically used with edrophonium (0.51.5 mg/kg) as they both have similar onsets of action (27). Glycopyrrolate (0.0070.015 mg/kg) is used to counteract the cholinergic effects of neostigmine (0.040.07 mg/kg). Alternatively, 0.2 mg of glycopyrrolate (IV) for each 1.0 mg of neostigmine or 5.0 mg of pyridostigmine can be given (27).

    Clinicians should recognize that muscle weakness can occur when anticholinesterases are given to patients with full neu-romuscular recovery (2529). Clinical studies have demon-strated that administration of anticholinesterases in the setting of near complete neuromuscular recovery (TOF 0.9) resulted in a decrease in minute ventilation, upper airway muscle tone, and diaphragmatic function (28, 29).

    Newer drugs that selectively encapsulate aminosteroid non-depolarizing agents (sugammadex-a modified A-cyclodextrin) provide more rapid reversal of aminosteroid NMBAs (3034). Sugammadex acts by forming tight complexes with these agents. Plasma steroidal neuromuscular blockers are quickly removed. The subsequent drop in plasma aminosteroid agent concentration produces a concentration gradient that allows for more aminosteroid to move from the NMJ into plasma. This remaining NMBA rapidly forms inactive complexes with the available plasma sugammadex (30). Sugammadex is cur-rently not available in the United States (but is available in Europe), nor has it been tested in the critically ill (3034). However, it may allow for the use of steroidal agents for urgent/emergent intubation in those patients in which succinylcho-line administration is contraindicated (see urgent/emergent intubation section). Furthermore, it may serve as a life-saving measure in those patients who receive aminosteroidal agents for intubation and who cannot be ventilated or intubated. Ongoing investigations may also address whether sugamma-dex can be used to reliably mitigate aminosteroid agent-asso-ciated anaphylaxis (35). Studies are warranted to validate its clinical use in the ICU.

    MonitoringRepeated clinical, qualitative, and quantitative evaluation of the depth of neuromuscular blockade may result in a reduction of NMBA dose and a subsequent decrease risk of complica-tions from residual neuromuscular blockade (36). Intensivists may look to achieve the absence of spontaneous ventilation to improve patient-ventilator synchrony. Clinical signs (such as the presence of spontaneous ventilation, eye opening, sus-tained hand grip, leg lift, or sustained 5-s head lift), used alone to determine the extent of muscle relaxation, are poor deter-minants in predicting the presence of residual neuromuscular blockade (37). In addition, these signs are not useful in those patients who are uncooperative.

    Peripheral nerve stimulators can facilitate appropriate titra-tion of NMBAs (94). TOF monitoring is the primary qualitative

    method used for assessment of neuromuscular blockade and involves four supramaximal electrical impulses every 0.5 sec-onds applied to the ulnar, facial, or posterior tibial nerve (1, 13). Figure 2 depicts a peripheral nerve stimulator eliciting a TOF response. Stimulation of these nerves will produce four visualized muscle twitches from the associated muscles (typi-cally muscles of the thumb or eye) that are innervated by the nerves. Fade of the twitch response occurs with increasing neuromuscular blockade (37) (Fig. 2C). Evaluation of fade can be performed by comparing the strength (TOF ratio) of the fourth twitch to that of the first twitch (25, 37).

    Several factors play a role in the assessment of TOF moni-toring. The loss of electrode skin adhesion, incorrect electrode placement, peripheral edema, and hypothermia (dampens peripheral stimulator twitch responses) can all contribute to inaccuracies in interpreting the TOF response (37, 38). TOF monitoring coupled with clinical evaluation has been shown to be insensitive for predicting residual neuromuscular weakness (25, 37). Patients with residual neuromuscular weakness can

    On/Off

    DBS Sustainedtetanus

    Train- of-Four

    SingleTwitch

    - mA+

    2a. TOF pattern (0.5 seconds each)

    2b. Response in absence of NMB

    2c. Response with partial NMB

    Figure 2. Depicts a peripheral nerve stimulator applied to stimulate the ulnar nerve and the train-of-four (TOF) stimulation pattern with no neu-romuscular blockade (NMB) and partial blockade. A, TOF pattern (0.5 s each). B, Response in absence of NMB. C, Response with partial NMB. DBS = double burst suppression.

  • Concise Definitive Review

    Critical Care Medicine www.ccmjournal.org 1337

    present with difficulty swallowing, rapid shallow breathing, nasal flaring, and overuse of accessory muscles (39). Studies suggest that residual neuromuscular blockade may lead to postoperative complications, including aspiration, atelectasis, pneumonia, and acute respiratory failure (25, 3740).

    Quantitative neuromuscular monitoring allows clini-cians to both stimulate the peripheral nerve and quantify the response to nerve stimulation (37). These devices are not rou-tinely used in the ICU. Acceleromyography is available and has been used in the operating room (37). Acceleromyography modules have been integrated in some bedside monitoring systems and can be used in the operating room and ICU. Figure 3 depicts how this monitor is applied on the patient. This device may help reduce signs and symptoms of residual neuromuscular weakness along with respiratory complica-tions (hypoxemia and airway obstruction) (41, 42). Research is needed regarding quantitative monitoring and its utiliza-tion in the ICU.

    A limited number of studies demonstrate a significant clini-cal benefit of neuromuscular monitoring for ICU patients (43, 44). An early open-label study using qualitative neuromuscular monitoring and adequate sedation showed that ICU patients did not experience prolonged paralysis, weakness, or other neuromuscular sequelae (45). Rudis et al (46) observed that neuromuscular monitoring decreased the amount of drug per hour, reduced the total drug used, and improved recovery time from neuromuscular blockade. Follow-up studies using a similar comparison with either atracurium or cisatracurium did not find this positive effect (47, 48). A more recent double-blinded randomized trial in a pediatric ICU found that neuro-muscular monitoring did not eliminate prolonged recovery in patients on vecuronium infusions (49). Trials are warranted to determine which patient populations will derive benefits from neuromuscular monitoring.

    NMBAs AND ASSOCIATED COMPLICATIONS

    ICU-Acquired WeaknessThe causes for ICU-acquired skeletal muscle weakness (ICU-AW) remain controversial. ICU-AW is classified as critical ill-ness polyneuropathy, critical illness myopathy, or critical illness neuromyopathy based on different electrophysiologic test find-ings (43, 50, 51). Proposed mechanisms for ICU-AW include disturbances in the microcirculation, protein malnutrition, systemic inflammation, and prolonged immobility (43, 50).

    More than 40 case reports have implicated NMBAs as a possi-ble cause for ICU-AW (43, 52, 53). However, there is a paucity of well-designed trials that confirm this relationship (43). The exist-ing studies examine a heterogeneous critically ill patient popu-lation and are methodologically compromised by variability in data collection (43). Other proposed risk factors, such as the use of high-dose corticosteroids, hyperglycemia, variation in age among studied populations, amount of sedation used, and type and duration of NMBAs administration, were not consistently controlled (43, 50, 51). Inconsistent use of electrophysiologic and manual muscle testing complicates interpretation of the pub-lished studies. Some investigators have suggested that these tests do not correlate with muscle function or outcome (43, 50, 51).

    Two recent trials did not identify an increased prevalence of ICU-AW in ARDS patients administered a 48-hour cisatracu-rium infusion (54, 55). Future investigations should examine the impact of other factors associated with ICU-AW, such as corticosteroids, sedation use, type and duration of NMBAs, or immobility. Trials should be performed on additional popu-lations (non-ARDS) to investigate whether limited use of NMBAs is associated with ICU-AW (43).

    Awareness During Induced ParalysisA recent case series reported 11 ICU patients who had been aware during pharmacologic paralysis (56). The common themes reported were thoughts of life and death, weird dreams, fear, and resisting being tied down. It is imperative to achieve a deep sedative state with sedatives and analgesics prior to the initiation of an NMBA to mitigate the risk of awareness. The authors suggest that strict adherence to sedation guidelines and protocols may facilitate appropriate titration of drugs while reducing awareness during paralysis. They also recom-mended that monitors should be developed to help clinicians appropriately titrate NMBAs, sedatives, and hypnotics as seda-tion titration protocols are not adequate enough to eliminate awareness (5661).

    No continuous bedside monitor has been universally used to alert clinicians of the potential for awareness. Processed electro-encephalogram models, such as the bispectral index (BIS mon-itor-Aspect Medical Systems, Covidien, Mansfield, MA), have been introduced into ICUs that may quantitatively monitor the level of consciousness (62). A recent ICU study suggested a satisfactory correlation between BIS values and validated sedation scales (63), whereas others have refuted it (6265). Well-designed clinical trials are needed to validate a correlation between sedation scales and continuous bedside monitors.

    Piezoelectric sensor

    Figure 3. Illustration of how the hand-held acceleromyography device is applied to a patient. This device stimulates the ulnar nerve producing thumb movement. A piezoelectric sensor is fixed onto the thumb and senses the acceleration of thumb movement. This acceleration is then sensed by the displayed monitor.

  • Greenberg and Vender

    1338 www.ccmjournal.org May 2013 Volume 41 Number 5

    DVT and Corneal AbrasionsNMBA administration to ICU patients may lead to nosocomial complications, such as DVT and corneal abrasions. Immobil-ity achieved by paralysis increases stasis of flow leading to an increase in DVT rate (66). A recent study reported that the use of NMBAs is the strongest predictor for the development of ICU-related DVTs (67). The same trial reported that educa-tional initiatives to promote DVT prophylaxis (both mechani-cal and pharmacologic) significantly reduced the prevalence of DVTs (67). Closer neuromuscular monitoring coupled with protocols to guide titration of NMBAs may also contribute to reducing the DVT prevalence by reducing the time to recovery from neuromuscular blockade (67).

    NMBAs also increase the risk of corneal abrasions in criti-cally ill patients. Paralysis abolishes eyelid closure and the blink reflex, which may result in cornea drying, scarring, ulceration, and infection (66). The prevalence of corneal abrasions in the adult critically ill varies from 8% to 60%, depending on the study design (68). Prophylactic eye protection (i.e., artificial ointments, eye covers, or methylcellulose drops) may reduce the prevalence of this ophthalmic complication (68).

    AnaphylaxisAllergic reactions associated with NMBAs are typically IgE-mediated (69). The ammonium ion in many NMBAs is the likely allergic component, which is also seen in many com-mon household products. Patients may develop anaphylaxis after the first dose of NMBA due to cross reactivity with other inciting agent exposures. Seven consecutive large French sur-veys over an 18-year period suggested that NMBAs are the most frequent perioperative agents (more than sedatives, hypnotics, latex, antibiotics, and colloids) involved in allergic reactions (70, 71).

    A diagnostic approach to suspected cases of anaphylaxis may include the following: obtaining a detailed medical his-tory, observing clinical signs (under deep sedation-pulseless-ness, oxygen desaturation, bronchospasm, and difficulty in ventilating), performing mediator release assays at the time of the reaction (i.e., tryptase levels), quantifying specific IgE immediately or during the first 6 months after the event, and performing skin tests and other biologic assays, such as hista-mine release tests or basophil activation (69, 71). The mainstay for diagnosis is the combination of obtaining the appropri-ate history with skin testing. Skin prick (SPT) or intradermal testing (IDT) are typically performed within 6 weeks of the reaction. Skin tests for NMBA usually remain positive for years following the exposure. Control testing (with saline) should be performed to ensure an accurate result. Skin testing has a high reported sensitivity (9497%) (69). SPT is preferred when investigating an NMBA anaphylactic reaction, whereas IDT is recommended to investigate a cross reaction (69, 71).

    GuidelinesCritical Care Medicine published ICU guidelines in 2002 stat-ing that NMBAs should be used for an adult patient in an ICU to manage ventilation, manage increased ICP, treat muscle

    spasms, and decrease oxygen consumption only when all other means have been tried without success (Grade C recommen-dation) (72). Both clinical and TOF monitoring was also rec-ommended (Grade B) so that the least amount of drug is used for the shortest duration (72). Guidelines have been developed for the titration of sedation and analgesics (7376). Clinicians should evaluate the reason for (at least daily) and depth of neu-romuscular blockade (continuously) so that mobility may be achieved faster, which may decrease the prevalence of delirium, increase functional independence, and increase ventilator-free days (77, 78).

    NMBA ICU Applications: Urgent/Emergent IntubationSuccinylcholine (11.5 mg/kg) is typically used to establish control of the airway to reduce the risk of aspiration due to its rapid onset (3060 s) and short duration of action (510 min) (79, 81). A 2008 Cochrane Database meta-analysis (37 stud-ies included) suggested that succinylcholine administration was associated with superior intubating conditions when compared with rocuronium (82). When rocuronium (1.2 mg/kg) was used, it was associated with similar intubating con-ditions when compared with succinylcholine. However, suc-cinylcholine was still reported to be clinically superior due to its shorter duration of action (82). A more recent randomized controlled trial in the ICU compared rocuronium (0.6 mg/kg) with succinylcholine (83). This trial suggested that there was no difference between the two drugs with regard to intubating conditions, prevalence and severity of oxygen desaturations, and prevalence of failed intubation attempts (83). Succinyl-choline should be avoided in the following clinical conditions where its depolarizing effect can cause a substantial increase in extracellular potassium: sustained muscle weakness, prolonged immobility, massive trauma with significant muscle injury, severe abdominal infections, concomitant acidosis and severe hypovolemia, or burns. Patients with renal failure can develop an increase in plasma potassium (approximately 0.5 mEq/L1 mEq/L) with use of succinylcholine, and therefore, it is rela-tively contraindicated in these patients (7981).

    Nondepolarizing NMBAs are the alternatives to succinyl-choline for endotracheal intubation. High-dose rocuronium (11.2 mg/kg) is commonly used for rapid intubation due to its relative short onset (12 min) of action (79). However, the duration of action is much longer with this dose (approxi-mately 6080 min) when compared with the standard intubating dose of rocuronium 0.6 mg/kg (2035 min). Rocuronium has a significant longer duration of action when compared with succinylcholine, and therefore, it may not be the appropriate drug for an anticipated difficult intuba-tion or mask ventilation as hypoxemia may occur (79, 82). Caution should also be used in patients with aminosteroid allergies (anaphylaxis has been reported with rocuronium) (71). Cisatracurium may be advantageous for intubation in patients with both liver and renal dysfunction. However, its onset of action is longer than rocuronium and its duration of action is comparable or somewhat longer than rocuronium. This does not make it an ideal drug for securing the airway

  • Concise Definitive Review

    Critical Care Medicine www.ccmjournal.org 1339

    emergently (47, 81). A recent study investigated the use of NMBAs for emergent endotracheal intubation in 566 patients from two major tertiary care centers (84). The use of NMBAs was associated with an improvement in intubating condi-tions and a reduction in both the prevalence of hypoxemia and procedure-related complications (aspiration, traumatic intubation, esophageal intubation, dental injury, and endo-bronchial intubation) (84).

    ALI/ARDSALI/ARDS is managed by treating the underlying cause of respiratory failure while reducing the risk of ventilator-induced lung injury (6, 85, 86). Induction of paralysis may increase chest wall compliance, eliminate patient-ventilator dyssynchrony, facilitate lung recruitment, reduce inflam-matory mediator release, decrease lung hyperinflation, and reduce oxygen consumption (albeit controversial) (6, 87). At least eight clinical trials have assessed the use of NMBAs for managing ALI/ARDS (Table 3) (1). Two small observational trials in ALI/ARDS patients prior to the year 2000 reported no effect on oxygenation when using a pancuronium bolus (88, 89). A large retrospective study by Arroliga et al (3) in

    2005 noted that 13% of patients were administered NMBAs for at least 1 day. NMBA use was associated with prolonged duration of mechanical ventilation, ICU length of stay, and an increase in mortality (3). A small randomized crossover trial in severe sepsis patients reported that oxygen consumption or delivery was not improved in patients randomized to receive NMBAs (90).

    Short-term use of cisatracurium in patients with ALI/ARDS has been associated with beneficial outcomes. Two small ran-domized controlled trials using cisatracurium in patients with ARDS demonstrated an improvement in oxygenation (91, 92). Another randomized trial in ARDS patients demonstrated that the early use of cisatracurium for 48 hours improved oxy-genation and reduced concentrations of pulmonary inflam-matory mediators (54). A larger multicenter trial in patients with severe ARDS (PaO

    2/FIO

    2 < 150 mm Hg) reported that

    early administration of cisatracurium for 48 hours increased adjusted 90-day survival, ventilator-free days, organ failurefree days, and reduced barotrauma (55). Further studies are required to define the appropriate combination of sedation and NMBAs and to identify when NMBA use will improve outcomes in patients with ALI/ARDS.

    TABLE 3. Use of Neuromuscular Blocking Agents for Acute Respiratory Distress Syndrome in the ICU

    Citation Study Design NMBA Regimen Conclusions

    Papazian et al (54) RCT (n = 340), severe ARDS Cisatracurium infusion 48 hr vs placebo

    Improved adjusted 90-d survival (p = 0.04), increased ventilator-free days (p = 0.03), increased organ dysfunctionfree days (p = 0.01), decreased barotrauma (p = 0.03)

    Arroliga et al (7) Retrospective (n = 549), ALI/ARDS

    High PEEP group, 45% had NMBA; low PEEP group, 33% had NMBA

    No difference in 60-d mortality or length of mechanical ventilation

    Forel et al (55) RCT (n = 36), ARDS Cisatracurium infusion 48 hr vs placebo

    Significant reduction in pulmonary/systemic cytokines (p = 0.005, p = 0.04) and increased oxygenation (p < 0.001)

    Arroliga et al (3) Retrospective (n = 5183), patient with mechanical ventilation 12 hr

    13% of patients given NMBA

    Significant increase in ICU mortality (p < 0.001), duration of mechanical ventilation (p < 0.001), and ICU length of stay (p < 0.001)

    Gainnier et al (91) RCT (n = 56), severe ARDS Cisatracurium infusion 48 hr vs placebo

    Improvement in oxygenation (p = 0.021), decrease in FIO2 (p < 0.001), and decrease in PEEP (p = 0.036)

    Lagneau et al (92) RCT (n = 102), ARDS Cisatracurium (2-hr infusion) used train-of-four to titrate NMBA

    Increase in oxygenation in groups with NMBA (p = 0.0014)

    Conti et al (89) Prospective, open label (n = 13); ALI/ARDS

    1 bolus-pancuronium No difference in oxygenation

    Bishop (88) Prospective, open label (n = 9); ALI/ARDS

    1 bolus-pancuronium No difference in oxygenation

    NMBA = neuromuscular blocking agent, RCT = randomized controlled trial, ALI = acute lung injury, ARDS = acute respiratory distress syndrome, PEEP = positive end-expiratory pressure.

  • Greenberg and Vender

    1340 www.ccmjournal.org May 2013 Volume 41 Number 5

    Status AsthmaticusNMBAs are used sparingly for patients with status asthmati-cus to minimize ventilator asynchrony, lung hyperinflation, and barotrauma (44). Table 4 presents several trials examin-ing NMBA use in patients with status asthmaticus. ICU-AW is reported in this population (93) and appears to be more commonly associated with the use of concomitant high-dose steroids (9498). A recent retrospective study in mechani-cally ventilated patients with status asthmaticus suggested that patients who require deep sedation with persistent immobiliza-tion are still at risk for weakness, despite a decline in the dura-tion of induced paralysis (97). The risk of prolonged weakness may be reduced by using the minimal doses of sedation, corti-costeroids, and NMBAs required to achieve the desired effect and to withdraw these drugs as soon as possible (97, 98).

    Increased ICPNMBAs are typically reserved for patients with intracranial hypertension in whom sedation alone does not reduce ICP

    (99). Few studies have evaluated the use of NMBAs for the management of elevations in ICP due to coughing, suctioning, or movement (100107) (Table 5). Hsiang et al (100) suggested that while NMBAs may reduce mortality, they may increase severe disability. The routine use of NMBAs for elevated ICP does not appear to be indicated at this time (106, 107). Patients who receive extended neuromuscular blockade as a second tier therapy for elevated ICP can be monitored with peripheral nerve stimulation to potentially avoid unwanted side effects (see Monitoring section) (99).

    Increased IAPIntra-abdominal hypertension (IAH) is defined as a repeated elevation in IAP of at least 12 mm Hg (108110). ACS is defined as continued IAH of greater than 20 mm Hg with end-organ dysfunction or failure (109, 110). Reduced abdominal wall compliance due to third spaced fluids, tense abdominal closures, and pain or inadequate sedation can lead to further increases in IAPs and subsequent organ failure (110). NMBAs

    TABLE 4. Use of Neuromuscular Blocking Agents for Status Asthmaticus in ICU

    Author Study Design% of patient receiving NMBA Results

    Kesler et al (97) Retrospective (n = 170) 67% before 1995, 77% after 1995

    No statistically significant difference in weakness between NMBA group and sedation-only group

    Adnet et al (96) Retrospective (n = 118) 54% Higher prevalence of muscle weakness, longer mechanical ventilation, longer ICU stay, and increase prevalence of pneumonia in NMBA group

    Behbehani et al (130) Retrospective (n = 86) 35% Increase myopathies with NMBAs

    Leatherman et al (94) Retrospective (n = 107) 65% Increase prevalence of muscle weakness with steroids + NMBAs, but no increase muscle weakness with use of benzylisoquinoliniums

    Griffin et al (131) Case series (n = 3), reviewed (n = 18) case reports

    100% All patients received both steroids and steroidal NMBA and all patients developed severe muscle weakness

    NMBA = neuromuscular blocking agent.

    TABLE 5. The Use of Neuromuscular Blocking Agents in Neurointensive Care

    Author Study Design% of NMBA Used or Type of NMBA Results

    Schramm et al (103) Randomized controlled trial (n = 14), neurosurgery

    Cisatracurium or atracurium bolus

    Atracurium-transient decrease in ICP, MAP, CPP, CBF; cisatracurium, no effect on MAP, ICP, CPP, CBF

    Schramm et al (101) Prospective, open label (n = 24)

    Cisatracurium bolus No effect on MAP, ICP, CPP, CBF

    Prielipp et al (102) Prospective, open label (n = 6)

    Doxacurium bolus and infusion

    No effect on MAP, ICP, no prolonged weakness

    Hsiang et al (100) Retrospective (n = 514) traumatic brain injury

    NMBA used in < 50% patients

    Decrease mortality with NMBA but increase prevalence of severely disabled patients, pneumonia, and ICU stay

    NMBA = neuromuscular blocking agent, ICP = intracranial pressure, MAP = mean arterial pressure, CPP = cerebral perfusion pressure, CBF = cerebral blood flow.

  • Concise Definitive Review

    Critical Care Medicine www.ccmjournal.org 1341

    have been reported to improve elevated IAPs by reducing abdominal muscle tone (110113).

    Neuromuscular blockade may provide clinicians with more time to remove fluid or treat the underlying cause of IAH and avoid surgical decompression (113115). Table 6 presents a few studies examining the use of NMBA in the treatment of IAH. A small prospective trial suggested that a bolus dose of cisatracu-rium was effective in significantly decreasing mild elevations in IAP (111). A recent case report describing a patient undergoing adrenalectomy who had ACS reported that a prolonged NMBA infusion of 48 hours was effective in reducing IAPs and provid-ing a good recovery (114). It has been recommended (Grade 2C) by the International ACS Consensus Definitions Conference Committee that NMBAs can be used in selected patients to reduce mild-to-moderate elevations of IAP (109, 110).

    Therapeutic Hypothermia After Out-of-Hospital VF-Associated Cardiac ArrestIn 2002, two landmark trials demonstrated that using mild hypothermia (3234 C) for 1224 hours in unconscious patients after out-of-hospital VF cardiac arrest improved neu-rologic outcomes, with one trial suggesting a survival benefit (116, 117). This therapy has been validated in other random-ized studies and meta-analyses (8, 118127). NMBAs were primarily used to prevent shivering (8, 116, 117) in the two original trials (and others).

    The optimal combination of sedatives, analgesics, and paralytics during hypothermia after VF-associated cardiac arrest is unknown. Chamorro et al (127) performed a sys-tematic review of 44 studies (68 international ICUs, primar-ily European) examining intensivists preferences for the administration of sedatives, analgesics, and NMBAs during therapeutic hypothermia after VF cardiac arrest. A significant variability in the protocolized use of all medications for these patients was found. Fifty-four ICUs routinely used NMBAs to prevent shivering, whereas eight ICUs used NMBAs to treat shivering (127). Pancuronium was the primary agent used in 24 international ICUs surveyed, whereas cisatracurium was the second most common agent used. A minority of ICUs used neuromuscular monitoring to guide NMBA dosing and therapy (127). Studies are required to elucidate the optimal type, dose, and mode of administration of NMBAs, as well

    as optimal combination of sedatives and analgesics in this patient population (128, 129).

    CONCLUSIONIntensivists use NMBAs for a variety of clinical conditions. Constant review and exploration of pharmacology, dosing, drug interactions, and monitoring techniques may reduce the unwanted side effects of NMBAs. Adoption of a collabora-tive ICU team effort coupled with the use of evidence-based guidelines may improve patient outcomes when using NMBAs. These measures may ensure that NMBAs are used and moni-tored appropriately.

    REFERENCES 1. Warr J, Thiboutot Z, Rose L, et al: Current therapeutic uses, pharma-

    cology, and clinical considerations of neuromuscular blocking agents for critically ill adults. Ann Pharmacother 2011; 45:11161126

    2. Merriman HM: The techniques used to sedate ventilated patients. A survey of methods used in 34 ICUs in Great Britain. Intensive Care Med 1981; 7:217224

    3. Arroliga A, Frutos-Vivar F, Hall J, et al; International Mechanical Ven-tilation Study Group: Use of sedatives and neuromuscular blockers in a cohort of patients receiving mechanical ventilation. Chest 2005; 128:496506

    4. Mehta S, Burry L, Fischer S, et al; Canadian Critical Care Trials Group: Canadian survey of the use of sedatives, analgesics, and neuromuscular blocking agents in critically ill patients. Crit Care Med 2006; 34:374380

    5. Rhoney DH, Murray K: National survey of the use of sedating drugs, neuromuscular blocking agents and reversal agents in the intensive care unit. J Intensive Care 2003; 18:139145

    6. Raoof S, Goulet K, Esan A, et al: Severe hypoxemic respiratory failure: Part 2nonventilatory strategies. Chest 2010; 137:14371448

    7. Arroliga AC, Thompson BT, Ancukiewicz M, et al; Acute Respiratory Distress Syndrome Network: Use of sedatives, opioids, and neuro-muscular blocking agents in patients with acute lung injury and acute respiratory distress syndrome. Crit Care Med 2008; 36:10831088

    8. Bernard SA, Buist M: Induced hypothermia in critical care medicine: A review. Crit Care Med 2003; 31:20412051

    9. Fagerland MJ, Erikkson LI: Current concepts in neuromuscular trans-mission. Br J Anaesth 2009; 103:108114

    10. Jonsson M, Dabrowski M, Gurley DA, et al: Activation and inhibition of human muscular and neuronal nicotinic acetylcholine receptors by succinylcholine. Anesthesiology 2006; 104:724733

    11. Jonsson Fagerlund M, Dabrowski M, Eriksson LI: Pharmacological characteristics of the inhibition of nondepolarizing neuromuscular blocking agents at human adult muscle nicotinic acetylcholine recep-tor. Anesthesiology 2009; 110:12441252

    TABLE 6. Studies Involving Neuromuscular Blocking Agent and Intra-Abdominal Hypertension

    Author Study Design NMBAs Results

    Cheatham et al (132) Prospective observational study (n = 478)

    Used combined medical/surgical management algorithm which contained the use of NMBA

    Increased patient survival to hospital discharge from 50% to 72% (p = 0.15)

    Davies et al (114) Case report (n = 1) Atracurium infusion 48 hr Reduced IAP and provided good recovery with discharge at 12 d postoperatively

    De Waele et al (111) Case series (n = 10) Cisatracurium bolus 9/10 patients had reduction in IAP

    NMBA = neuromuscular blocking agent, IAP = intraabdominal pressure.

  • Greenberg and Vender

    1342 www.ccmjournal.org May 2013 Volume 41 Number 5

    12. McManus MC: Neuromuscular blockers in surgery and intensive care, part I. Am J Health Syst Pharm 2001; 58:22872299

    13. Miller R: Miller: Millers Anesthesia. Second Edition. Orlando, FL, Churchill Livingstone, 2009

    14. Murphy GS, Vender J: Neuromuscular blocking agents use and mis-use in the intensive care unit. Crit Care Clin 2001; 17:925942

    15. Smith BS, Yogaratnam D, Levasseur-Franklin KE, et al: Introduction to drug pharmacokinetics in the critically ill patient. Chest 2012; 141:13271336

    16. Caldwell JE, Heier T, Wright PM, et al: Temperature-dependent phar-macokinetics and pharmacodynamics of vecuronium. Anesthesiology 2000; 94:8392

    17. Smeulers NJ, Wierda JM, van den Broek L, et al: Effects of hypo-thermic cardiopulmonary bypass on the pharmacodynamics and pharmacokinetics of rocuronium. J Cardiothorac Vasc Anesth 1995; 9:700705

    18. Beaufort AM, Wierda JM, Belopavlovic M, et al: The influence of hypothermia (surface cooling) on the time-course of action and on the pharmacokinetics of rocuronium in humans. Eur J Anaesthesiol Suppl 1995; 11:95106

    19. Atherton DP, Hunter JM: Clinical pharmacokinetics of the newer neu-romuscular blocking drugs. Clin Pharmacokinet 1999; 36:169189

    20. Magorian T, Wood P, Caldwell J, et al: The pharmacokinetics and neuromuscular effects of rocuronium bromide in patients with liver disease. Anesth Analg 1995; 80:754759

    21. Szenohradszky J, Fisher DM, Segredo V, et al: Pharmacokinetics of rocuronium bromide (ORG 9426) in patients with normal renal func-tion or patients undergoing cadaver renal transplantation. Anesthesi-ology 1992; 77:899904

    22. Cooper RA, Maddineni VR, Mirakhur RK, et al: Time course of neuro-muscular effects and pharmacokinetics of rocuronium bromide (Org 9426) during isoflurane anaesthesia in patients with and without renal failure. Br J Anaesth 1993; 71:222226

    23. Boyd AH, Eastwood NB, Parker CJ, et al: Comparison of the phar-macodynamics and pharmacokinetics of an infusion of cis-atracurium (51W89) or atracurium in critically ill patients undergoing mechani-cal ventilation in an intensive therapy unit. Br J Anaesth 1996; 76:382388

    24. Tatro DS (Ed): Non-depolarizing muscle relaxants. In: Drug Interaction Facts. St Louis, MO, Wolters Kluwer, 2010, pp 12311253

    25. Murphy GS, Brull SJ: Residual neuromuscular block: Lessons unlearned. Part II: Methods to reduce the risk of residual weakness. Anesth Analg 2010; 111:129140

    26. Caldwell JE: Clinical limitations of acetylcholinesterase antagonists. J Crit Care 2009; 24:2128

    27. Bevan DR, Donati F, Kopman AF: Reversal of neuromuscular block-ade. Anesthesiology 1992; 77:785805

    28. Eikermann M, Zaremba S, Malhotra A, et al: Neostigmine but not sugammadex impairs upper airway dilator muscle activity and breath-ing. Br J Anaesth 2008; 101:344349

    29. Eikermann M, Fassbender P, Malhotra A, et al: Unwarranted adminis-tration of acetylcholinesterase inhibitors can impair genioglossus and diaphragm muscle function. Anesthesiology 2007; 107:621629

    30. de Boer HD, van Egmond J, van de Pol F, et al: Sugammadex, a new reversal agent for neuromuscular block induced by rocuronium in the anaesthetized Rhesus monkey. Br J Anaesth 2006; 96:473479

    31. Fuchs-Buder T, Meistelman C, Schreiber JU: Is sugammadex economi-cally viable for routine use. Curr Opin Anaesthesiol 2012; 25:217220

    32. Cammu G, De Kam PJ, Demeyer I, et al: Safety and tolerability of sin-gle intravenous doses of sugammadex administered simultaneously with rocuronium or vecuronium in healthy volunteers. Br J Anaesth 2008; 100:373379

    33. Naguib M, Brull SJ: Sugammadex: A novel selective relaxant binding agent. Expert Rev Clin Pharmacol 2009; 2:3753

    34. Akha AS, Rosa J 3rd, Jahr JS, et al: Sugammadex: Cyclodextrins, development of selective binding agents, pharmacology, clinical devel-opment, and future directions. Anesthesiol Clin 2010; 28:691708

    35. McDonnell MJ, Pavy TJ, Green LK, et al: Sugammadex in the man-agement of rocuronium induced anaphylaxis. Br J Anesth 2011; 106:199201

    36. Vender JS, Szokol J, Murphy GS, et al: Sedation, analgesia, neuro-muscular blockade in sepsis: An evidence based review. Crit Care Med 2004; 32:S554561

    37. Murphy GS, Brull SJ: Residual neuromuscular block: Lessons unlearned. Part I: Definitions, incidence, and adverse physiologic effects of residual neuromuscular blockade. Anesth Analg 2010; 111:120128

    38. Pino RM: Neuromuscular blockade studies of critically ill patients. Intensive Care Med 2002; 28:16951697

    39. Greenberg SB, Vender JS: Postoperative pulmonary complications. Curr Rev Clin Anesth 2011; 31:221232

    40. Sauer M, Stahn A, Soltesz S, et al: The influence of residual neuro-muscular blockade on the incidence of critical respiratory events. A randomized prospective, placebo controlled trial. European J Anes-thesiol 2011; 28:841848

    41. Murphy GS, Szokol JW, Marymont JH, et al: Intraoperative accelero-myographic monitoring reduces the risk of residual neuromuscular blockade and adverse respiratory events in the postanesthesia care unit. Anesthesiology 2008; 109:389398

    42. Murphy GS, Szokol JW, Avram MJ, et al: Intraoperative accelero-myography monitoring reduces symptoms of muscle weakness and improves quality of recovery in the early postoperative period. Anes-thesiology 2011; 115:946954

    43. Puthucheary Z, Rawal J, Ratnayake G, et al: Neuromuscular blockade and skeletal muscle weakness in critically ill patients: Time to rethink the evidence? Am J Respir Crit Care Med 2012; 185:911917

    44. Forel JM, Roch A, Papazian L: Paralytics in critical care: Not always the bad guy. Curr Opin Crit Care 2009; 15:5966

    45. Khuenl-Brady KS, Reitsttter B, Schlager A, et al: Long-term admin-istration of pancuronium and pipecuronium in the intensive care unit. Anesth Analg 1994; 78:10821086

    46. Rudis MI, Sikora CA, Angus E, et al: A prospective, randomized, con-trolled evaluation of peripheral nerve stimulation versus standard clini-cal dosing of neuromuscular blocking agents in critically ill patients. Crit Care Med 1997; 25:575583

    47. Strange C, Vaughan L, Franklin C, et al: Comparison of train-of-four and best clinical assessment during continuous paralysis. Am J Respir Crit Care Med 1997; 156:15561561

    48. Baumann MH, McAlpin BW, Brown K, et al: A prospective randomized comparison of train-of-four monitoring and clinical assessment during continuous ICU cisatracurium paralysis. Chest 2004; 126:12671273

    49. Burmester M, Mok Q: Randomised controlled trial comparing cisatra-curium and vecuronium infusions in a paediatric intensive care unit. Intensive Care Med 2005; 31:686692

    50. Stevens RD, Marshall SA, Cornblath DR, et al: A framework for diag-nosing and classifying intensive care unit-acquired weakness. Crit Care Med 2009; 37(10 Suppl):S299S308

    51. Coakley JH, Nagendran K, Yarwood GD, et al: Patterns of neurophysi-ological abnormality in prolonged critical illness. Intensive Care Med 1998; 24:801807

    52. MacFarlane IA, Rosenthal FD: Severe myopathy after status asthmati-cus. Lancet 1977; 2:615

    53. Schakman O, Gilson H, Thissen JP: Mechanisms of glucocorticoid-induced myopathy. J Endocrinol 2008; 197:110

    54. Papazian L, Forel JM, Gacouin A, et al; ACURASYS Study Investiga-tors: Neuromuscular blockers in early acute respiratory distress syn-drome. N Engl J Med 2010; 363:11071116

    55. Forel JM, Roch A, Marin V, et al: Neuromuscular blocking agents decrease inflammatory response in patients presenting with acute respiratory distress syndrome. Crit Care Med 2006; 34:27492757

    56. Ballard N, Robley L, Barrett D, et al: Patients recollections of ther-apeutic paralysis in the intensive care unit. Am J Crit Care 2006; 15:8694; quiz 95

    57. Loper KA, Butler S, Nessly M, et al: Paralyzed with pain: The need for education. Pain 1989; 37:315316

    58. Brook AD, Ahrens TS, Schaiff R, et al: Effect of a nursing-implemented sedation protocol on the duration of mechanical ventilation. Crit Care Med 1999; 27:26092615

    59. Kress JP, Pohlman AS, OConnor MF, et al: Daily interruption of seda-tive infusions in critically ill patients undergoing mechanical ventila-tion. N Engl J Med 2000; 342:14711477

  • Concise Definitive Review

    Critical Care Medicine www.ccmjournal.org 1343

    60. Girard TD, Kress JP, Fuchs BD, et al: Efficacy and safety of a paired sedation and ventilator weaning protocol for mechanically ventilated patients in intensive care (Awakening and Breathing Controlled trial): A randomised controlled trial. Lancet 2008; 371:126134

    61. Patel SB, Kress JP: Sedation and analgesia in the mechanically venti-lated patient. Am J Respir Crit Care Med 2012; 185:486497

    62. Tonner PH, Paris A, Scholz J: Monitoring consciousness in intensive care medicine. Best Pract Res Clin Anaesthesiol 2006; 20:191200

    63. Yaman F, Ozcan A, Kaymak C, et al: Assessment of correlation between bispectral index and four common sedation scales used in mechanically ventilated patients in ICU. Eur Rev Med Pharmacol Sci 2012; 16:660666

    64. Haenggi M, Ypparila-Wolters H, Buerki S, et al: Auditory event-related potentials, bispectral index, and entropy for the discrimination of dif-ferent levels of sedation in intensive care unit patients. Anesth Analg 2009; 109:807816

    65. LeBlanc JM, Dasta JF, Pruchnicki MC, et al: Bispectral index values, sedation agitation scores, and plasma concentrations of lorazepam in critically ill surgical patients. Am J Crit Care 2012; 21:99105

    66. Honiden S, Siegel MD: Analytic reviews: Managing the agitated patient in the ICU: Sedation, analgesia, and neuromuscular blockade. J Intensive Care Med 2010; 25:187204

    67. Boddi M, Barbani F, Abbate R, et al: Reduction in deep venous throm-bosis incidence in intensive care after a clinician education program. Thromb Hemost 2009; 8:121128

    68. Sorce LR, Hamilton SM, Gauvreau K, et al: Preventing corneal abra-sions in critically ill children receiving neuromuscular blockade: A ran-domized controlled trial. Pediatr Crit Care Med 2009; 10:171175

    69. Mertes PM, Tajima K, Regnier-Kimmoun MA, et al: Perioperative ana-phylaxis. Med Clin North Am 2010; 94:761789

    70. Mertes PM, Laxenaire MC: Anaphylatic and anaphylactoid reactions occurring during anesthesia in France. Seventh epidemiologic sur-vey (January 2001-December 2002). Ann Fr Anesth Reanim 2004; 23:11331143

    71. Dewachter P, Mouton-Faivre CM, Emala CW: Anaphylaxis and anes-thesia. Anesthesiology 2009; 111:11411150

    72. Murray MJ, Cowen J, Deblock H, et al: Clinical practice guidelines for sustained neuromuscular blockade in the critically ill adult. Crit Care Med 2002; 30;142156

    73. Bailey P, Thomsen GE, Spuhler VJ, et al: Early activity is feasible and safe in respiratory failure patients. Crit Care Med 2007; 35:139145

    74. Thomsen GE, Snow GL, Rodriguez L, et al: Patients with respira-tory failure increase ambulation after transfer to an intensive care unit where early activity is a priority. Crit Care Med 2008; 36:11191124

    75. Morris PE, Goad A, Thompson C, et al: Early intensive care unit mobil-ity therapy in the treatment of acute respiratory failure. Crit Care Med 2008; 36:22382243

    76. Schweickert WD, Pohlman MC, Pohlman AS, et al: Early physical and occupational therapy in mechanically ventilated, critically ill patients: A randomised controlled trial. Lancet 2009; 373:18741882

    77. Mascia MF, Koch M, Medicis JJ: Pharmacoeconomic impact of rational use guidelines on the provision of analgesia, sedation, and neuromuscular blockade in critical care. Crit Care Med 2000; 28:23002306

    78. Chanques G, Barbotte JS, Violet S, et al: Impact of systemic evalu-ation of pain and agitation in an intensive care unit. Crit Care Med 2006; 34:16911699

    79. Reynolds SF, Heffner J: Airway management of the critically ill patient: Rapid-sequence intubation. Chest 2005; 127:13971412

    80. El-Orbany M, Connolly LA: Rapid sequence induction and intuba-tion: Current controversy. Anesth Analg 2010; 110:13181325

    81. Booij LH: Is succinylcholine appropriate or obsolete in the intensive care unit? Crit Care 2001; 5:245246

    82. Perry J, Lee J, Sillberg VA, et al: Rocuronium versus succinylcholine for rapid sequence induction intubation. Cochrane Database Syst Rev 2008; 2:CD002788

    83. Marsch SC, Steiner L, Bucher L, et al: Succinylcholine versus rocuronium for rapid sequence intubation in intensive care: A pro-spective, randomized trial. Crit Care 2011; 15:R199R208

    84. Wilcox SR, Bittner EA, Elmer J, et al: Neuromuscular blocking agent administration for emergent tracheal intubation is associated with decreased prevalence of procedure-related complications. Crit Care Med 2012; 40:18081813

    85. Rubenfeld GD, Herridge MS: Epidemiology and outcomes of acute lung injury. Chest 2007; 131:554562

    86. Zambon M, Vincent JL: Mortality rates for patients with acute lung injury/ARDS have decreased over time. Chest 2008; 133:11201127

    87. Bennett S, Hurford WE: When should sedation or neuromuscu-lar blockade be used during mechanical ventilation? Respir Care 2011; 56:168176

    88. Bishop MJ: Hemodynamic and gas exchange effects of pancuronium bromide in sedated patients with respiratory failure. Anesthesiology 1984; 60:369371

    89. Conti G, Vilardi V, Rocco M, et al: Paralysis has no effect on chest wall and respiratory system mechanics of mechanically ventilated, sedated patients. Intensive Care Med 1995; 21:808812

    90. Freebairn RC, Derrick J, Gomersall CD, et al: Oxygen delivery, oxy-gen consumption, and gastric intramucosal pH are not improved by a computer-controlled, closed-loop, vecuronium infusion in severe sepsis and septic shock. Crit Care Med 1997; 25:7277

    91. Gainnier M, Roch A, Forel JM, et al: Effect of neuromuscular block-ing agents on gas exchange in patients presenting with acute respi-ratory distress syndrome. Crit Care Med 2004; 32:113119

    92. Lagneau F, Dhonneur G, Plaud B, et al: A comparison of two depths of prolonged neuromuscular blockade induced by cisatracurium in mechanically ventilated critically ill patients. Intensive Care Med 2002; 28:17351741

    93. Appleton R, Kinsella J: Intensive care unit-acquired weakness. Con-tin Educ Anaesth Crit Care Pain 2012; 12:6266

    94. Leatherman JW, Fluegel WL, David WS, et al: Muscle weakness in mechanically ventilated patients with severe asthma. Am J Respir Crit Care Med 1996; 153:16861690

    95. Goh AY, Chan PW: Acute myopathy after status asthmaticus: Ste-roids, myorelaxants or carbon dioxide? Respirology 1999; 4:9799

    96. Adnet F, Dhissi G, Borron SW, et al: Complication profiles of adult asthmatics requiring paralysis during mechanical ventilation. Inten-sive Care Med 2001; 27:17291736

    97. Kesler SM, Sprenkle MD, David WS, et al: Severe weakness com-plicating status asthmaticus despite minimal duration of neuromus-cular paralysis. Intensive Care Med 2009; 35:157160

    98. Oddo M, Feihl F, Schaller MD, et al: Management of mechanical ventilation in acute severe asthma: Practical aspects. Intensive Care Med 2006; 32:501510

    99. Ohlinger MJ, Rhoney DH: Neuromuscular blocking agents in the neurosurgical intensive care unit. Surg Neurol 1998; 49:217221

    100. Hsiang JK, Chesnut RM, Crisp CB, et al: Early, routine paralysis for intracranial pressure control in severe head injury: Is it necessary? Crit Care Med 1994; 22:14711476

    101. Schramm WM, Jesenko R, Bartunek A, et al: Effects of cisatra-curium on cerebral and cardiovascular hemodynamics in patients with severe brain injury. Acta Anaesthesiol Scand 1997; 41: 13191323

    102. Prielipp RC, Robinson JC, Wilson JA, et al: Dose response, recovery, and cost of doxacurium as a continuous infusion in neu-rosurgical intensive care unit patients. Crit Care Med 1997; 25: 12361241

    103. Schramm WM, Papousek A, Michalek-Sauberer A, et al: The cere-bral and cardiovascular effects of cisatracurium and atracurium in neurosurgical patients. Anesth Analg 1998; 86:123127

    104. Kerr ME, Sereika SM, Orndoff P, et al: Effect of neuromuscular blockers and opiates on the cerebrovascular response to endotra-cheal suctioning in adults with severe head injuries. Am J Crit Care 1998; 7:205217

    105. Juul N, Morris GF, Marshall SB, et al: Neuromuscular blocking agents in neurointensive care. Acta Neurochir Suppl 2000; 76:467470

    106. Rangel-Castillo L, Gopinath S, Robertson CS: Management of intracranial hypertension. Neurol Clin 2008; 26:521541

  • Greenberg and Vender

    1344 www.ccmjournal.org May 2013 Volume 41 Number 5

    107. Brain Trauma Foundation, American Association of Neurological Surgeons (AANS), Congress of Neurological Surgeons (CNS), AANS/CNS Joint Section on Neurotrauma and Critical Care: Guidelines for the management of traumatic brain injury. J Neu-rotrauma 2007; 24:S71S76

    108. Cheatham ML: Intraabdominal pressure monitoring during fluid resuscitation. Curr Opin Crit Care 2008; 14:327333

    109. Malbrain ML, CheathamML, Kirkpatrick A, et al: Results from the conference of experts on intra-abdominal hypertension and abdomi-nal compartment syndrome. Part I: Definitions. Intensive Care Med 2006; 32:17221732

    110. Cheatham ML, Malbrain MLNG, Kirkpatrick A, et al: Results from the conference of experts on intra-abdominal hypertension and abdominal compartment syndrome. Part II: Recommendations. Intensive Care Med 2007; 33:951962

    111. De Waele J, Delaet I, Hoste E, et al. The effect of neuromuscular blockers on intraabdominal pressure. Crit Care Med 2006; 34:A70

    112. Mertens zur Borg IR, Verbrugge SJ, Kolkman KA: Anesthetic consid-erations in abdominal compartment syndrome. In: Abdominal Com-partment Syndrome. Ivatury RR, Cheatham ML, Malbrain MLNG, et al (Eds). Landes Biomedical, Georgetown, 2006, pp 254265

    113. An G, West MA: Abdominal compartment syndrome: A concise clinical review. Crit Care Med 2008; 36:13041310

    114. Davies J, Aghahoseini A, Crawford J, et al: To close or not to close? Treat-ment of abdominal compartment syndrome by neuromuscular blockade without laparostomy. Ann R Coll Surg Engl 2010; 92:W8W9

    115. Luckianow GM, Ellis M, Governale D, et al: Abdominal compartment syndrome: Risk factors, diagnosis, and current therapy. Crit Care Res Pract 2012; 908169:18

    116. Bernard SA, Gray TW, Buist MD, et al: Treatment of comatose sur-vivors of out-of-hospital cardiac arrest with induced hypothermia. N Engl J Med 2002; 346:557563

    117. The Hypothermia After Cardiac Arrest Study Group: Mild therapeu-tic hypothermia to improve the neurologic outcome after cardiac arrest. N Engl J Med 2002; 346:549556

    118. Polderman KH: Application of therapeutic hypothermia in the ICU: Opportunities and pitfalls of a promising treatment modality. Part 1: Indications and evidence. Intensive Care Med 2004; 30: 556575

    119. Holzer M, Bernard SA, Hachimi-Idrissi S, et al; Collaborative Group on Induced Hypothermia for Neuroprotection After Cardiac Arrest: Hypothermia for neuroprotection after cardiac arrest: Systematic review and individual patient data meta-analysis. Crit Care Med 2005; 33:414418

    120. Ramani R: Hypothermia for brain protection and resuscitation. Curr Opin Anaesthesiol 2006; 19:487491

    121. Alzaga AG, Cerdan M, Varon J: Therapeutic hypothermia. Resusci-tation 2006; 70:369380

    122. Rincon F, Mayer SA: Therapeutic hypothermia for brain injury after cardiac arrest. Semin Neurol 2006; 26:387395

    123. Howes D, Green R, Gray S, et al; Canadian Association of Emer-gency Physicians: Evidence for the use of hypothermia after cardiac arrest. CJEM 2006; 8:109115

    124. Cheung KW, Green RS, Magee KD: Systematic review of random-ized controlled trials of therapeutic hypothermia as a neuroprotec-tant in post cardiac arrest patients. CJEM 2006; 8:329337

    125. Holzer M, Behringer W: Therapeutic hypothermia after cardiac arrest and myocardial infarction. Best Pract Res Clin Anaesthesiol 2008; 22:711728

    126. Schneider A, Bttiger BW, Popp E: Cerebral resuscitation after car-diocirculatory arrest. Anesth Analg 2009; 108:971979

    127. Chamorro C, Borrallo JM, Romera MA, et al: Anesthesia and anal-gesia protocol during therapeutic hypothermia after cardiac arrest: A systematic review. Anesth Analg 2010; 110:13281335

    128. Heier T, Caldwell JE: Impact of hypothermia on the response to neu-romuscular blocking drugs. Anesthesiology 2006; 104:10701080

    129. Prielipp RC, Coursin DB: Applied pharmacology of common neu-romuscular blocking agents in critical care. New Horiz 1994; 2:3447

    130. Behbehani NA, Al-Mane F, Dyachkova Y, et al: Myopathy following mechanical ventilation for severe acute asthma: The role of muscle relaxants and corticosteroids. Chest 1999; 115:16271631

    131. Griffin D, Fairman N, Coursin D, et al: Acute myopathy during treat-ment of status asthmaticus with corticosteroids and steroidal mus-cle relaxants. Chest 1992; 102:510514

    132. Cheatham ML, Safcsak K: Is the evolving management of intra-abdominal hypertension and abdominal compartment syndrome improving survival? Crit Care Med 2010; 38:402407