Back To Search Results

Neuromuscular Blocking Agents

Editor: David A. Layer Updated: 6/8/2024 1:51:26 PM

Indications

The widespread adoption of neuromuscular blocking agents (NMBAs), or drugs (NMBDs), significantly impacted the field of anesthesia. Before the introduction of NMBAs, anesthesia was induced and maintained with intravenous and inhalational agents. Afterward, anesthesia was redefined to include the triad of hypnosis, analgesia, and muscle relaxation.[1][2]

History

Native peoples in South America historically coated arrows and darts used for hunting with a substance called "curare." Spanish explorers, or Conquistadors, returned home from expeditions to South America, telling tales of a "flying death." In 1562, a writer in the court of King Ferdinand and Queen Isabella of Spain was the first to document anecdotal evidence of these poisoned projectiles in a book titled De Orbe Novo.[3] Sir Walter Raleigh, a British explorer, described the use of poisoned arrows in modern-day Venezuela in his 1848 book titled Discovery of the Large, Rich, and Beautiful Empire of Guiana. The word "ourari" was used by one of Sir Walter's lieutenants.[4][5] Edward Bancroft, another pioneer, brought samples of crude curare from South America back to the Old World.

Sir Benjamin Brodie performed the first medical experiments with curare. He demonstrated that small animals injected with curare stopped breathing but could be kept alive by inflating their lungs with bellows. He concluded that curare causes the cessation of breathing by paralyzing the respiratory muscles.

Charles Waterton, a manager of a large sugar plantation estate in South America, became interested in the effects of a substance the native population called "wourali" (a local term for curare). In 1814, Waterton demonstrated these effects on 3 donkeys. The first donkey was injected in the shoulder and died soon afterward. The second donkey had a tourniquet applied to the foreleg and was injected with curare distal to the tourniquet. This donkey lived while the tourniquet was in place but died soon after the tourniquet's removal. The third donkey appeared to be dead after injection but was resuscitated after using bellows to respirate on the animal's behalf. Charles Waterton's experiment confirmed the paralytic effect of curare.

Another milestone in the development of NMBAs involves the work of French physiologist Claude Bernard (1813-1878). Bernard demonstrated that when curare is injected into a frog leg, the muscle in the leg does not contract when the innervating nerve is stimulated. However, contraction does occur with direct stimulation to the muscle itself. This experiment showed that the curare acts on the neuromuscular junction.

After studying curare, neurologist Walter Freeman hypothesized that administering curare to induce temporary paralysis could provide relief of muscle spasms and rigidity in patients with multiple sclerosis (MS.) Freeman shared his hypothesis with Richard Gill, a patient with MS, who brought 25 lbs (11 kg) of raw curare from Ecuador. The raw curare was delivered to Squibb & Sons, who attempted to derive an effective antidote to curare. In 1942, Squibb & Sons scientists Wintersteiner and Dutcher isolated the alkaloid d-tubocurarine. Soon after, AH Holladay (also working for Squibb & Sons) developed a standardized commercial preparation of curare named Intocostrin.[6]

Neuropsychiatrist E. Bennett, a pioneer in the field of convulsive shock therapy, used curare to minimize the risk of spinal fracture during these procedures. Bennet presented a film demonstrating this use of curare at the 91st Congress of the American Medical Association. Lewis Wright, an employee of Squibb & Sons, was present at this Congress and was intrigued by Bennett's film. He donated some Intocostrin to E.A. Rovenstine of New York University, who passed the medication on to E.M. Papper, one of his residents. The administration of Intocostrin to 2 patients caused a cessation of breathing; Papper and colleagues manually ventilated both patients overnight. 

In 1942, Harold Randall Griffith and his resident Enid Johnson administered curare to a young patient undergoing an appendectomy at the Homeopathic Hospital in Montreal.[7][8][9] This is considered the first major step towards using NMBA for muscle relaxation during anesthesia.

Several synthetic NMBAs were developed during the 1940s, 50s, and 60s. Gallamine was the first synthetic NMBA administered clinically. Scientists later developed suxamethonium (succinylcholine), pancuronium, vecuronium, atracurium, and rocuronium.

FDA-Approved Indications

Indications for NMBA administration include:

  • Endotracheal intubation [10]
  • Therapeutic hypothermia after cardiac arrest
  • Acute respiratory distress syndrome [11][12]
  • Elevated intraabdominal pressure
  • Elevated intracranial pressure
  • Status asthmaticus [13]
  • Prevention of patient-ventilator asynchrony in patients on mechanical ventilation [14]
  • Muscular relaxation for a surgical procedure
  • Adjunct therapy for patients undergoing electroconvulsive therapy
  • Rapid sequence intubation [15]

Mechanism of Action

Register For Free And Read The Full Article
Get the answers you need instantly with the StatPearls Clinical Decision Support tool. StatPearls spent the last decade developing the largest and most updated Point-of Care resource ever developed. Earn CME/CE by searching and reading articles.
  • Dropdown arrow Search engine and full access to all medical articles
  • Dropdown arrow 10 free questions in your specialty
  • Dropdown arrow Free CME/CE Activities
  • Dropdown arrow Free daily question in your email
  • Dropdown arrow Save favorite articles to your dashboard
  • Dropdown arrow Emails offering discounts

Learn more about a Subscription to StatPearls Point-of-Care

Mechanism of Action

NMBAs act at the neuromuscular junction (NMJ), which consists of 3 parts:

  • Presynaptic nerve terminal
  • Synaptic cleft
  • Postsynaptic nicotinic receptors

When an electric impulse transmits along the motor neuron, it causes the release of acetylcholine (ACh) from the presynaptic membrane, which travels across the synaptic cleft and acts on nicotinic receptors on the postsynaptic membrane, causing muscle contraction.

Presynaptic Nerve Terminal

The presynaptic nerve terminal consists of motor neurons originating from the ventral horn of the spinal cord. The motor neuron loses its myelin sheath as it embeds in muscle tissue and secretes trophic and growth factors. The nicotinic acetylcholine receptors (nAChRs) are located on the surface of the presynaptic nerve terminal membrane.[16]

Acetylcholine is stored in vesicles that are readily releasable or gathered in a reserve pool. Readily-released ACh activates sodium channels in the presynaptic membrane, which activate voltage-dependent calcium channels (P-type fast), allowing an inward movement of calcium into the cytoplasm of the motor neuron.[16]

The Synaptic Cleft

The synaptic cleft is the space between the presynaptic and postsynaptic membranes, typically measuring about 50 nm across. Several biologically active substances interact in the cleft to promote and enhance nerve impulse transmission. These substances include acetylcholine esterase (AChE), lipoprotein receptor protein 4 (LRP4), and agrin. A glycoprotein binds LRP4 in the cleft, activating muscle-specific tyrosine kinase (MuSK), which helps differentiate acetylcholine receptors.

The Postsynaptic Membrane

This membrane comprises multiple shoulders densely populated with acetylcholine receptors (AChRs) and voltage-gated sodium channels. This density is possible due to rapsyn and other proteins serving as anchors, enabling a neurotransmitter signal to elicit sufficient depolarization for muscle contraction.[17]

AChR receptors exist in 2 forms:

Mature (adult) junctional receptors are pentametric proteins with 5 subunits. They have high conductivity, remain open for a short period, and have a half-life of approximately 14 days.

Immature (fetal) junctional receptors are primarily found in fetal tissue but can increase in conditions like sepsis, burns, and upper and lower motor neuron diseases. They have a pentameric structure, a half-life of 24 hours, and appear within 24 hours after injury. These receptors are resistant to non-depolarizing neuromuscular blocking agents but are sensitive to succinylcholine. Stimulation of these receptors can cause potassium efflux, predisposing to hyperkalemia.

Depolarizing Neuromuscular Blocking Agents

Depolarizing NMBAs (eg, succinylcholine) act on receptors at the motor endplate of the neuromuscular junction (NMJ), causing depolarization of the membrane; this induces a refractory period. These drugs have an onset of action of 1 minute and a duration of 6 minutes and are rapidly metabolized by plasma butyrylcholinesterase.[18] The continued disruption of ACh-mediated effects causes muscular fasciculation and twitching. Succinylcholine, or suxamethonium, is the only depolarizing NMBA used clinically.

Nondepolarizing Neuromuscular Blocking Agents

Nondepolarizing NMDAs prevent acetylcholine from binding to the motor plate at the NMJ by competing for the ACh binding site on the α subunit of nicotinic receptors. As the concentration of non-depolarizing NMBAs at the junction increases relative to ACh, a neuromuscular blockade becomes established.[19]

There are 2 major structural classes of non-depolarizing NMBAs:

Aminosteroids Neuromuscular Blockers 

  • Vecuronium
  • Pancuronium
  • Rocuronium

Benzylisoquinolinium Neuromuscular Blockers

  • Benzylisoquinolinium
  • Mivacurium
  • Atracurium
  • Cis–atracurium [19] 
  • Gantacurium [20] (not FDA-approved)

Based on duration of action, NMBDs may be classified as:

  • Short-acting (eg, mivacurium, succinylcholine)
  • Intermediate-acting (eg, vecuronium, rocuronium, atracurium)
  • Long-acting (eg, pancuronium, gallamine, tubocurarine) [21]

Administration

Available Dosage Forms and Strengths

Neuromuscular blocking agents (NMBAs) are most effective when administered intravenously or intramuscularly. They are poorly absorbed if administered orally.[22] The administration route is determined by the patient's clinical condition, desired action speed, and clinical effect duration. NMBA administration is typically intravenous through boluses or continuous infusion. Continuous infusion is usually preferred in the ICU, where prolonged paralysis may be necessary, or in the OR, where a surgical procedure requires significant time.

Adult Dosage

NMBA dosing is based on ideal body weight to prevent overdosing or prolonged paralysis.[23] Adequate sedation and analgesia should be established before NMBA administration. The Society of Critical Care Medicine Clinical Practice Guidelines for Rapid Sequence Intubation recommends administering a neuromuscular blocking agent with a sedative-hypnotic induction agent in critically ill adult patients. Five studies have demonstrated improved first-pass intubation success (FPS) and reduced respiratory arrest, cardiovascular collapse, and vomiting/aspiration occurrences with NMBA use. Due to their widespread availability, NMBAs should be reserved for airway management in critically ill patients when an induction agent is planned to induce unconsciousness.[15] These dosages serve as general guidelines and may require modification depending on individual patient characteristics and use in the ICU vs surgery. For precise dosing, providers should refer to the FDA-approved labeling for each medication and carefully assess the patient's clinical situation. This approach ensures the safe and tailored administration of drugs, aligning with best healthcare practices.

  • Succinylcholine: 0.6 mg/kg, Maintenance: 0.04 to 0.07 mg/kg 

  • Atracurium: Initial bolus: 0.4 to 0.5 mg/kg, Maintenance: 0.08 to 0.1 mg/kg 

  • Cisatracurium: Initial bolus: 0.15 to 0.2 mg/kg, Maintenance: 0.03 mg/kg IV 

  • Rocuronium: Intubation (RSI): 0.6 to 1.2 mg/kg, Maintenance: 0.01 to 0.012 mg/kg/min

  • Vecuronium: Initial bolus: 0.08 to 0.1 mg/kg, Maintenance:0.8 to 1.2 μg/kg/min

Specific Patient Populations

Hepatic impairment: Liver disease can reduce plasma cholinesterase activity, leading to prolonged neuromuscular blockade after succinylcholine administration.[24]

Renal impairment: Atracurium is the preferred neuromuscular blocking agent for chronic kidney disease (CKD) due to its metabolism via ester hydrolysis and Hofmann degradation, which are independent of renal function. Cisatracurium is also suitable, as it is metabolized by Hofmann degradation, though its clearance is reduced by 13% in CKD. Vecuronium and rocuronium undergo significant renal excretion, leading to prolonged action and reduced clearance in CKD; they require careful monitoring. Pancuronium should be avoided due to decreased clearance.[25] 

Pregnancy considerations: Succinylcholine is preferred for its rapid elimination and minimal transfer to breast milk during pregnancy and postpartum. Other non-depolarizing muscle relaxants, such as pipecuronium and vecuronium, are considered safe alternatives for a caesarian section due to their limited placental transfer and minimal fetal effects. Rocuronium is widely used during pregnancy, as it has a shorter onset and is reversed with sugammadex. Atracurium and cisatracurium are safe options with low placental transfer and minimal cardiovascular effects. Mivacurium, while having the shortest duration, should be used cautiously in patients at risk of eclampsia. Depolarizing and non-depolarizing neuromuscular blocking drugs are ionized and water-soluble, preventing placental transfer at clinically relevant doses. Rocuronium and succinylcholine are classified by the FDA as previous Pregnancy Categories B and C, respectively. These drugs do not cause neonatal skeletal muscle weakness or paralysis. Pregnancy decreases plasma cholinesterase concentration due to increased plasma volume, affecting the onset and duration of these drugs. Therefore, careful monitoring of neuromuscular function and cautious use of reversal agents are essential to prevent rapid increases in circulating acetylcholine, which may induce uterine contractions.[26][27]

Breastfeeding considerations: Non-depolarizing neuromuscular blocking drugs are quaternary ammonium compounds with limited lipid solubility and oral absorption. They are primarily ionized at physiological pH, making them unlikely to pass into breast milk in significant quantities. Due to its ionization at physiological pH and poor oral absorption, succinylcholine is unlikely to be present in notable amounts in breast milk. Breastfeeding can resume once the woman has recovered from anesthesia. Limited clinical data is available on rocuronium during breastfeeding. Rocuronium's short action, high polarity, and poor oral absorption suggest minimal transfer to breast milk and a low risk to the infant.[28] Neostigmine is a quaternary ammonium with a half-life of 15 to 30 minutes that may be transferred to breast milk in nonsignificant quantities. Although excreted in breast milk based on animal studies, sugammadex is a large, polar molecule with minimal likelihood of oral absorption by the infant. Sugammadex is considered safe for use during breastfeeding.[29]

Pediatric patients: Pediatric patients exhibit different responses to neuromuscular blocking agents than adults due to developmental changes in neuromuscular transmission and body composition. Infants are particularly sensitive to non-depolarizing neuromuscular blocking agents because of deficient acetylcholine levels in developing motor nerves. However, this sensitivity is primarily offset by the distribution of these drugs into a larger extracellular fluid volume. When performing rapid sequence intubation in pediatric patients, succinylcholine or rocuronium should be considered (due to their rapid onset), with sugammadex available for reversal.[30]

Older patients: Older patients are more susceptible to postoperative residual neuromuscular blockade (PRNB) and its associated complications, including airway obstruction, hypoxemic events, muscle weakness symptoms, postoperative pulmonary complications, and extended stays in the PACU and hospital. In these patients, PRNB was linked to higher rates of adverse events.[31]

Adverse Effects

The different NMBA types have various adverse effects associated with their use.

Depolarizing NMBAs

Succinylcholine administration correlates with a significant elevation in serum potassium levels. Therefore, succinylcholine should be avoided for patients with chronic kidney disease, rhabdomyolysis, burns, or crush injuries. Elevated potassium levels can lead to fatal arrhythmias.

Succinylcholine can cause bradycardia, especially in pediatric patients. Activation of the nicotinic receptor can stimulate an associated muscarinic receptor, resulting in bradycardia. Administering succinylcholine with atropine or glycopyrrolate can mitigate this effect.

Succinylcholine can cause increased intracranial and intraocular pressure. The administration of adequate age-appropriate sedation in conjunction with succinylcholine can minimize this unwanted adverse effect.[18]

Succinylcholine can also cause malignant hyperthermia, a pharmacogenetic disorder triggered by the concurrent administration of volatile inhalation anesthetics and succinylcholine. Malignant hyperthermia clinically presents as hypercarbia, metabolic acidosis, hyperventilation, hyperthermia, and rhabdomyolysis. This condition is associated with RYR1 and CACNA1S gene mutations.[32]

Other adverse effects of succinylcholine include jaw rigidity, hypersalivation, and hypersensitivity reactions.

Nondepolarizing NMBAs

Benzylisoquinolinium Derivatives

Benzylisoquinolinium, mivacurium, atracurium, cisatracurium, and doxacurium can stimulate histamine release, causing bronchospasm, hypotension, and tachycardia due to peripheral vasodilation.

Laudanosine, a metabolite of atracurium, can accumulate in the central nervous system (CNS) and cause seizures.[33]

Amino Steroids

Prolonged infusion of vecuronium, pancuronium, or rocuronium with concurrent steroids can cause muscular weakness, a condition called "critical illness polyneuropathy."[34]

Potentiating Conditions

Some conditions can prolong the effects of neuromuscular blocking agents:

  • Hypothermia
  • Metabolic derangements
  • Hypercalcemia
  • Hypermagnesemia
  • Hypokalemia
  • Hypothermia
  • Respiratory acidosis
  • Metabolic alkalosis [35]

Drug-Drug Interactions

  • The administration of aminoglycosides (eg, neomycin, gentamicin, tobramycin) or clindamycin potentiates neuromuscular blockade and can enhance the blockade effect induced by rocuronium.[36]
  • Inhaled anesthetic agents act at the neuromuscular junction to potentiate neuromuscular blockade in a dose-dependent manner. Desflurane, sevoflurane, and isoflurane can enhance the effect of non-depolarizing neuromuscular blocking agents.[37]
  • The concentration-dependent modulatory effect of magnesium enhances and prolongs the action of non-depolarizing NMBAs.[38]
  • Medications like calcium channel blockers, lithium, cyclosporine, and anti-arrhythmic drugs such as procainamide and quinidine can increase sensitivity to neuromuscular blocking agents.[39]

Contraindications

Patients with a history of a severe allergic reaction to an NMBA or anaphylaxis should not take NMBAs. Pancuronium and vecuronium undergo deacetylation in the liver; these drugs should be administered to patients with liver failure cautiously. NMBAs are among the most common allergens responsible for hypersensitivity reactions during anesthesia, accounting for 70% of cases.[40]

Pseudocholinesterase deficiency impairs the body's ability to metabolize succinylcholine. The dibucaine inhibition test aids in diagnosing this condition by measuring enzyme activity inhibition after administering dibucaine; higher inhibition percentages indicate greater deficiency severity. The dibucaine number refers to the inhibition percentage.[2]

Box Warning

Succinylcholine can trigger rhabdomyolysis, a rare but critical complication in pediatric anesthesia. In young patients with undiagnosed Duchenne muscular dystrophy (DMD), this condition causes hyperkalemia, increasing the risk of life-threatening ventricular dysrhythmias and cardiac arrest. Management focuses on aggressive interventions for hyperkalemia, including the administration of intravenous calcium, bicarbonate, and glucose with insulin. To minimize the risk of rhabdomyolysis, succinylcholine use in pediatrics should be reserved for emergencies requiring immediate airway control.[41]

Warning and Precautions

  • Rocuronium and vecuronium are excreted via the hepatobiliary system and should be avoided in patients with liver failure if an alternative agent is available.
  • Pancuronium, doxacurium, and pipecuronium should be avoided in patients with kidney failure as these drugs are excreted renally.
  • Atracurium and cisatracurium may be used in patients with kidney or liver failure. These NMDAs get eliminated by a unique process called Hoffman elimination, a spontaneous degradation process independent of liver or kidney function.[42]
  • The extravasation of neuromuscular blocking agents can cause unpredictable neuromuscular blockades. Absorption from the subcutaneous depot is slower and influenced by lipid solubility, protein binding, and local tissue perfusion. The extravasation site affects pharmacokinetics, as faster onset and recovery are expected in more vascularized regions. Conditions such as diabetes and atherosclerosis can further influence absorption.[43]
  • Caution should be exercised for patients with myasthenia gravis due to their increased sensitivity to neuromuscular blockers. Reversal of rocuronium or vecuronium is achieved with sugammadex administration.[44]
  • Per the American Society of Anesthesiologists (ASA) guidelines for obstructive sleep apnea, a complete reversal of neuromuscular block must be confirmed before extubation.[45]

Monitoring

Patients on NMDAs are usually undergoing treatment in the intensive care unit. Monitoring parameters for these patients include pulse oximetry for oxygen saturation and continuous end-tidal CO2 (ETCO2). A subsequent elevation in carbon dioxide may suggest developing malignant hyperthermia.

Assessment of neuromuscular blockage involves measuring the "train-of-4" ratio. Typically, the ulnar, median, or facial nerves are stimulated to monitor the effect of neuromuscular blockade. Four electric impulses are delivered at a frequency of 2 Hz. The fourth-to-first twitch ratio is used to assess the blockade effect. The disappearance of the fourth twitch signifies a blockage of 75%, the disappearance of the third twitch signifies a blockage of 85%, the second twitch for 90%, and if only the first twitch is observable, the blockade is near 100%.[46][47][48] The American Society of Anesthesiologists 2023 guidelines recommend quantitative neuromuscular monitoring at the adductor pollicis muscle and confirmed recovery of the "train-of-4" to ≥90% before proceeding with extubation.[49]

The bispectral index monitor (BIS) is a type of quantitative electroencephalography (EEG) used to assess sedation of patients on continuous infusion of NMBAs using bi-spectrality and time domain.[50] In patients who are deeply sedated or under general anesthesia, the BIS is sensitive to changes in the patient's electromyogram (EMG).[51][52][53]

Toxicity

Signs and Symptoms of Toxicity

NMBA dosing should be based on ideal body weight. Excessive NMBA dosing results in paralysis for longer than required and may present with prolonged muscular weakness, decreased respiratory drive, and apnea. Certain conditions mentioned above can prolong the effect of NMBA. Inadequate recovery from neuromuscular blocking drugs is associated with adverse outcomes, including pneumonia, upper airway obstruction, reintubation, atelectasis, and prolonged stay in the postanesthesia care unit (PACU).

Antagonism of Neuromuscular Blockade

Residual neuromuscular blockade is defined as a "train-of-4" (TOF) ratio <0.9 at the adductor pollicis muscle, making it essential to confirm recovery at this site. Precise measurement of the blockade effect aids in selecting the appropriate antagonist drug and determining its dose. If intraoperative monitoring was performed using the eye muscles, the adductor pollicis muscle should be used to confirm the TOF ratio before administering an antagonist. Antagonist dosages are based on adductor pollicis responses, and FDA-approved sugammadex dosages do not apply to corrugator supercilii monitoring.

According to ASA guidelines, sugammadex is recommended for the neuromuscular blockade induced by vecuronium or rocuronium. Sugammadex encapsulates the neuromuscular blocking agent, rendering it inactive. Sugammadex dosing is based on actual body weight (not ideal) and is not recommended for patients with a creatinine clearance (CrCl) <30 mL/min. 

Selecting neostigmine or sugammadex is determined by the likelihood of effective reversal, the type of neuromuscular blocker, blockade depth, and antagonist efficacy. Proper use of antagonists and monitoring is crucial to prevent residual blockade.

Successful antagonism with neostigmine is best achieved when significant spontaneous recovery occurs, specifically at a TOF count of 4 twitches. Studies have demonstrated that administering neostigmine after observing a fourth twitch is associated with better outcomes than lower counts. However, effective NBMA reversal is only confirmed if the fourth twitch is strong. Neostigmine may be used for deeper blocks with an understanding of longer recovery times.

For an initial TOF ratio of 0.4, neostigmine administration achieves a TOF ratio of ≥0.9 within 10 minutes. For a TOF ratio of 0.4 to <0.9, neostigmine should not exceed a 40 µg/kg dose. Neostigmine at 30 µg/kg is recommended for minimal blockade. Higher doses can cause weakness if the TOF ratio is ≥0.9. Neostigmine reaches its maximum effect within 10 minutes; if a TOF ratio ≥0.9 is not observed in this timeframe, providers can wait for further recovery or administer sugammadex or additional neostigmine (no more than 50 µg/kg). 

In the absence of quantitative monitoring and given a TOF count of 4, 15 to 30 µg/kg of neostigmine may be administered, after which providers must wait 10 minutes before extubation. With quantitative monitoring, extubation can occur at a TOF ratio ≥0.9. 

The FDA-approved sugammadex dosages are also based on train-of-4 counts. These guidelines recommend 2 mg/kg for a TOF count of 2 (or ratio <0.9), 4 mg/kg for a TOF count of 1 (or a post-tetanic count of 1), and 16 mg/kg for immediate reversal after 1.2 mg/kg rocuronium administration. Women using hormonal contraceptives require contraception for 7 days after sugammadex administration.

Both neostigmine and sugammadex are effective at a TOF ratio of 0.5. Adverse effects of sugammadex and neostigmine (with glycopyrrolate) are similar, with no significant differences in the incidence of anaphylaxis, bradycardia, tachycardia, postoperative nausea, or vomiting.[49] 

Management of Extravasation

If extravasation is suspected, a new intravenous line for safe anesthesia should be secured promptly. After an accidental subcutaneous injection, a prolonged and unpredictable duration of neuromuscular blockade should be presumed, especially with additional intravenous doses of NMBDs. Quantitative monitoring to detect ongoing block risk should be regularly performed, with TOF monitoring ideally beginning at anesthesia induction to assess block onset. Before administering additional intravenous NMBA doses, the TOF should be measured to align with best practices for anesthesia. Due to potentially delayed absorption from subcutaneous tissue, spontaneous patient recovery may be observed, or active blockade may be further stabilized. Prolonged ventilation and sedation may be necessary post-surgery.

If an increase in TOF count or ratio is observed, sugammadex or neostigmine can reverse the effect of NMBAs. Sugammadex is preferred for aminosteroid NMBDs due to its pharmacological profile, neutralizing capability, lack of muscarinic effects, less residual paralysis, and longer half-life elimination than neostigmine. Sugammadex rarely causes perioperative anaphylaxis. Extubation is safe to perform when the TOF ratio exceeds 0.9. Post-extubation monitoring should include regular neuromuscular block measurement, full ASA monitoring, and a minimum PACU stay of 4 hours, particularly with long-acting NMBAs. Additional sugammadex may be given to reverse aminosteroid NMBD administration if the TOF ratio declines during the PACU observation period. Additional doses must not be given if neostigmine inadequately reverses a non-aminosteroid blockade. Instead, the patient should be observed in the ICU under prolonged sedation and ventilation.[43]

Enhancing Healthcare Team Outcomes

After a surgical procedure is completed or when a patient is weaning towards extubation, NMBAs may be reversed pharmacologically to prevent adverse effects and facilitate quick extubation. Traditionally, neostigmine is used to reverse the effect of NMBAs. Neostigmine's mechanism of action involves the inhibition of acetylcholinesterase (AChE), the enzyme responsible for the breakdown of acetylcholine (ACh). Elevated levels of ACh will compete with the NMBA and stimulate the nicotinic receptors at the neuromuscular junction, enhancing signal transmission.

Recent advances in anesthesia have introduced sugammadex, a cyclodextrin that selectively binds to NMBAs in the plasma. The encapsulation process rapidly nullifies the effect of the NMBA as it is unavailable to act at the neuromuscular junction.[54]

Sugammadex produces a safe and quick reversal of commonly used NMBAs like rocuronium, vecuronium, and pancuronium. Sugammadex can quickly reverse both moderate and deep neuromuscular blockade (NMB).[55][56][57]

With the introduction of sugammadex, deeper and longer NMB may be maintained during surgical procedures without fear of prolonged recovery. Deep NMB improves working conditions for surgeons with less insufflation pressure required during laparoscopic surgery.[58][59][60][61] 

NMBAs should be administered only by adequately trained professionals (eg, anesthesiologists, certified registered nurse anesthetists (CRNA), intensivists, and emergency physicians). The administration of NMBAs should not occur without personnel and facilities for resuscitation and life support, and an antagonist must be readily available. The pharmacist must ensure that dosage is appropriate for the clinical scenario and report any disparities. Respiratory therapists are essential in implementing weaning protocols for mechanical ventilation and ensuring lung-protective ventilation.[62] An interprofessional team approach and open communication between clinicians (MDs, DOs, NPs, PAs), CRNAs, respiratory therapists, pharmacists, and nurses is necessary to optimize patient outcomes related to neuromuscular blocking agents.

References


[1]

Tosh P, Rajan S. Safety and Efficacy of Dexmedetomidine versus Propofol Infusion as an Adjunct to Transtracheal Block in Ensuring Patient Immobility During General Anesthesia without the Use of Muscle Relaxants. Anesthesia, essays and researches. 2019 Oct-Dec:13(4):683-687. doi: 10.4103/aer.AER_134_19. Epub 2019 Dec 16     [PubMed PMID: 32009715]


[2]

Hsieh CY, Tan H, Huang HF, Huang TY, Wu CW, Chang PY, Lu DV, Lu IC. Optimization of Intraoperative Neural Monitoring of the Recurrent Laryngeal Nerve in Thyroid Surgery. Medicina (Kaunas, Lithuania). 2022 Mar 30:58(4):. doi: 10.3390/medicina58040495. Epub 2022 Mar 30     [PubMed PMID: 35454334]


[3]

Sykes K. Harold Griffith Memorial Lecture. The Griffith legacy. Canadian journal of anaesthesia = Journal canadien d'anesthesie. 1993 Apr:40(4):365-74     [PubMed PMID: 8485797]


[4]

Crul JF. Relaxant drugs: from native drugs to the selective agents of today. Acta anaesthesiologica Scandinavica. 1982 Aug:26(4):409-15     [PubMed PMID: 6181643]


[5]

Birmingham AT. Fifth W.D.M. Paton Memorial Lecture. Waterton and Wouralia. British journal of pharmacology. 1999 Apr:126(8):1685-9     [PubMed PMID: 10372809]

Level 3 (low-level) evidence

[6]

COHNBERG RE. Stimulation of the central nervous system by curare (intocostrin). The Journal of laboratory and clinical medicine. 1946 Aug:31():866-77     [PubMed PMID: 20995236]


[7]

Gillies D, Wynands JE. Harold Randall Griffith. The pioneer of the use of muscle relaxants in anaesthesia. British journal of anaesthesia. 1986 Sep:58(9):943-5     [PubMed PMID: 3530290]


[8]

Kyle RA, Shampo MA. Harold R. Griffith--introduction of muscle relaxants to anesthesia. Mayo Clinic proceedings. 1992 Mar:67(3):237     [PubMed PMID: 1545590]


[9]

. . :():     [PubMed PMID: 20988929]


[10]

deBacker J, Hart N, Fan E. Neuromuscular Blockade in the 21st Century Management of the Critically Ill Patient. Chest. 2017 Mar:151(3):697-706. doi: 10.1016/j.chest.2016.10.040. Epub 2016 Nov 3     [PubMed PMID: 27818334]


[11]

Wei XB, Wang ZH, Liao XL, Guo WX, Qin TH, Wang SH. Role of Neuromuscular Blocking Agents in Acute Respiratory Distress Syndrome: An Updated Meta-Analysis of Randomized Controlled Trials. Frontiers in pharmacology. 2019:10():1637. doi: 10.3389/fphar.2019.01637. Epub 2020 Jan 29     [PubMed PMID: 32063852]

Level 1 (high-level) evidence

[12]

Qadir N, Sahetya S, Munshi L, Summers C, Abrams D, Beitler J, Bellani G, Brower RG, Burry L, Chen JT, Hodgson C, Hough CL, Lamontagne F, Law A, Papazian L, Pham T, Rubin E, Siuba M, Telias I, Patolia S, Chaudhuri D, Walkey A, Rochwerg B, Fan E. An Update on Management of Adult Patients with Acute Respiratory Distress Syndrome: An Official American Thoracic Society Clinical Practice Guideline. American journal of respiratory and critical care medicine. 2024 Jan 1:209(1):24-36. doi: 10.1164/rccm.202311-2011ST. Epub     [PubMed PMID: 38032683]

Level 1 (high-level) evidence

[13]

Iavarone IG, Al-Husinat L, Vélez-Páez JL, Robba C, Silva PL, Rocco PRM, Battaglini D. Management of Neuromuscular Blocking Agents in Critically Ill Patients with Lung Diseases. Journal of clinical medicine. 2024 Feb 19:13(4):. doi: 10.3390/jcm13041182. Epub 2024 Feb 19     [PubMed PMID: 38398494]


[14]

Bailey JM. Management of Patient-Ventilator Asynchrony. Anesthesiology. 2021 Apr 1:134(4):629-636. doi: 10.1097/ALN.0000000000003704. Epub     [PubMed PMID: 33592102]


[15]

Acquisto NM, Mosier JM, Bittner EA, Patanwala AE, Hirsch KG, Hargwood P, Oropello JM, Bodkin RP, Groth CM, Kaucher KA, Slampak-Cindric AA, Manno EM, Mayer SA, Peterson LN, Fulmer J, Galton C, Bleck TP, Chase K, Heffner AC, Gunnerson KJ, Boling B, Murray MJ. Society of Critical Care Medicine Clinical Practice Guidelines for Rapid Sequence Intubation in the Critically Ill Adult Patient: Executive Summary. Critical care medicine. 2023 Oct 1:51(10):1407-1410. doi: 10.1097/CCM.0000000000005999. Epub 2023 Sep 14     [PubMed PMID: 37707378]

Level 1 (high-level) evidence

[16]

Fagerlund MJ, Eriksson LI. Current concepts in neuromuscular transmission. British journal of anaesthesia. 2009 Jul:103(1):108-14. doi: 10.1093/bja/aep150. Epub     [PubMed PMID: 19546202]


[17]

Burden SJ. SnapShot: Neuromuscular Junction. Cell. 2011 Mar 4:144(5):826-826.e1. doi: 10.1016/j.cell.2011.02.037. Epub     [PubMed PMID: 21376240]

Level 3 (low-level) evidence

[18]

Hager HH, Burns B. Succinylcholine Chloride. StatPearls. 2024 Jan:():     [PubMed PMID: 29763160]


[19]

Sparr HJ, Beaufort TM, Fuchs-Buder T. Newer neuromuscular blocking agents: how do they compare with established agents? Drugs. 2001:61(7):919-42     [PubMed PMID: 11434449]

Level 3 (low-level) evidence

[20]

Naguib M, Brull SJ. Update on neuromuscular pharmacology. Current opinion in anaesthesiology. 2009 Aug:22(4):483-90. doi: 10.1097/ACO.0b013e32832b8cff. Epub     [PubMed PMID: 19384229]

Level 3 (low-level) evidence

[21]

Raghavendra T. Neuromuscular blocking drugs: discovery and development. Journal of the Royal Society of Medicine. 2002 Jul:95(7):363-7     [PubMed PMID: 12091515]


[22]

Buck ML, Reed MD. Use of nondepolarizing neuromuscular blocking agents in mechanically ventilated patients. Clinical pharmacy. 1991 Jan:10(1):32-48     [PubMed PMID: 1825627]


[23]

Ingrande J, Lemmens HJ. Dose adjustment of anaesthetics in the morbidly obese. British journal of anaesthesia. 2010 Dec:105 Suppl 1():i16-23. doi: 10.1093/bja/aeq312. Epub     [PubMed PMID: 21148651]


[24]

Kiamanesh D, Rumley J, Moitra VK. Monitoring and managing hepatic disease in anaesthesia. British journal of anaesthesia. 2013 Dec:111 Suppl 1():i50-61. doi: 10.1093/bja/aet378. Epub     [PubMed PMID: 24335399]


[25]

Chowdhury SR, McLure HA. Chronic kidney disease and anaesthesia. BJA education. 2022 Aug:22(8):321-328. doi: 10.1016/j.bjae.2022.03.005. Epub 2022 Jun 15     [PubMed PMID: 36097571]


[26]

Radkowski P, Jacewicz M, Podhorodecka K. The Use of Muscle Relaxants in Pregnancy and Puerperium Period. International journal of general medicine. 2023:16():859-864. doi: 10.2147/IJGM.S393885. Epub 2023 Mar 3     [PubMed PMID: 36895512]


[27]

Shin J. Anesthetic Management of the Pregnant Patient: Part 2. Anesthesia progress. 2021 Jun 1:68(2):119-127. doi: 10.2344/anpr-68-02-12. Epub     [PubMed PMID: 34185861]


[28]

. Rocuronium. Drugs and Lactation Database (LactMed®). 2006:():     [PubMed PMID: 29999741]


[29]

Mitchell J, Jones W, Winkley E, Kinsella SM. Guideline on anaesthesia and sedation in breastfeeding women 2020: Guideline from the Association of Anaesthetists. Anaesthesia. 2020 Nov:75(11):1482-1493. doi: 10.1111/anae.15179. Epub 2020 Aug 1     [PubMed PMID: 32737881]


[30]

Hussain SY, Panjiar P, Jain D, Khanooja S, Batt KM. Current practice of Rapid Sequence Induction (RSI) in pediatric anesthesia: A survey from India. Journal of anaesthesiology, clinical pharmacology. 2023 Jan-Mar:39(1):88-97. doi: 10.4103/joacp.joacp_172_21. Epub 2022 Mar 4     [PubMed PMID: 37250269]

Level 3 (low-level) evidence

[31]

Murphy GS,Szokol JW,Avram MJ,Greenberg SB,Shear TD,Vender JS,Parikh KN,Patel SS,Patel A, Residual Neuromuscular Block in the Elderly: Incidence and Clinical Implications. Anesthesiology. 2015 Dec;     [PubMed PMID: 26448469]


[32]

Rosenberg H, Pollock N, Schiemann A, Bulger T, Stowell K. Malignant hyperthermia: a review. Orphanet journal of rare diseases. 2015 Aug 4:10():93. doi: 10.1186/s13023-015-0310-1. Epub 2015 Aug 4     [PubMed PMID: 26238698]


[33]

D'Souza RS, Porter BR, Johnson RL. Nondepolarizing Paralytics. StatPearls. 2024 Jan:():     [PubMed PMID: 30137795]


[34]

Murray MJ, Brull SJ, Bolton CF. Brief review: Nondepolarizing neuromuscular blocking drugs and critical illness myopathy. Canadian journal of anaesthesia = Journal canadien d'anesthesie. 2006 Nov:53(11):1148-56     [PubMed PMID: 17079642]


[35]

Waud BE, Waud DR. Interaction of calcium and potassium with neuromuscular blocking agents. British journal of anaesthesia. 1980 Sep:52(9):863-6     [PubMed PMID: 7437224]

Level 3 (low-level) evidence

[36]

Lee JH, Lee SI, Chung CJ, Lee JH, Lee SC, Choi SR, Oh JN, Bae JY. The synergistic effect of gentamicin and clindamycin on rocuronium-induced neuromuscular blockade. Korean journal of anesthesiology. 2013 Feb:64(2):143-51. doi: 10.4097/kjae.2013.64.2.143. Epub 2013 Feb 15     [PubMed PMID: 23459675]


[37]

Paul M, Fokt RM, Kindler CH, Dipp NC, Yost CS. Characterization of the interactions between volatile anesthetics and neuromuscular blockers at the muscle nicotinic acetylcholine receptor. Anesthesia and analgesia. 2002 Aug:95(2):362-7, table of contents     [PubMed PMID: 12145052]


[38]

Cho CK, Sung TY, Choi SJ, Choi HR, Kim YB, Lee JU, Yang HS. The effect of magnesium sulfate concentration on the effective concentration of rocuronium, and sugammadex-mediated reversal, in isolated left phrenic nerve hemi-diaphragm preparations from the rat. Korean journal of anesthesiology. 2018 Oct:71(5):401-406. doi: 10.4097/kja.d.17.27150. Epub 2018 May 30     [PubMed PMID: 29843202]


[39]

Gisselmann G, Alisch D, Welbers-Joop B, Hatt H. Effects of Quinine, Quinidine and Chloroquine on Human Muscle Nicotinic Acetylcholine Receptors. Frontiers in pharmacology. 2018:9():1339. doi: 10.3389/fphar.2018.01339. Epub 2018 Nov 20     [PubMed PMID: 30515099]


[40]

Dejoux A, de Chaisemartin L, Bruhns P, Longrois D, Gouel-Chéron A. Neuromuscular blocking agent induced hypersensitivity reaction exploration: an update. European journal of anaesthesiology. 2023 Feb 1:40(2):95-104. doi: 10.1097/EJA.0000000000001765. Epub 2022 Oct 18     [PubMed PMID: 36301083]


[41]

Bohringer C, Moua H, Liu H. Is There Still a Role for Succinylcholine in Contemporary Clinical Practice? Translational perioperative and pain medicine. 2019:6(4):129-135. doi: 10.31480/2330-4871/100. Epub 2019 Sep 12     [PubMed PMID: 33834087]


[42]

Agoston S, Vandenbrom RH, Wierda JM. Clinical pharmacokinetics of neuromuscular blocking drugs. Clinical pharmacokinetics. 1992 Feb:22(2):94-115     [PubMed PMID: 1551294]


[43]

Nietvelt F, Van Herreweghe I, Godschalx V, Soetens F. Extravascular injection of neuromuscular blocking drugs: A systematic review of current evidence and management. European journal of anaesthesiology. 2024 May 1:41(5):367-373. doi: 10.1097/EJA.0000000000001967. Epub 2024 Feb 27     [PubMed PMID: 38410855]

Level 1 (high-level) evidence

[44]

Carron M, De Cassai A, Linassi F. Sugammadex in the management of myasthenic patients undergoing surgery: beyond expectations. Annals of translational medicine. 2019 Dec:7(Suppl 8):S307. doi: 10.21037/atm.2019.10.35. Epub     [PubMed PMID: 32016026]


[45]

American Society of Anesthesiologists Task Force on Perioperative Management of patients with obstructive sleep apnea, Practice guidelines for the perioperative management of patients with obstructive sleep apnea: an updated report by the American Society of Anesthesiologists Task Force on Perioperative Management of patients with obstructive sleep apnea. Anesthesiology. 2014 Feb;     [PubMed PMID: 24346178]

Level 1 (high-level) evidence

[46]

Srivastava A, Hunter JM. Reversal of neuromuscular block. British journal of anaesthesia. 2009 Jul:103(1):115-29. doi: 10.1093/bja/aep093. Epub 2009 May 24     [PubMed PMID: 19468024]

Level 3 (low-level) evidence

[47]

Naguib M, Kopman AF, Lien CA, Hunter JM, Lopez A, Brull SJ. A survey of current management of neuromuscular block in the United States and Europe. Anesthesia and analgesia. 2010 Jul:111(1):110-9. doi: 10.1213/ANE.0b013e3181c07428. Epub 2009 Nov 12     [PubMed PMID: 19910616]

Level 3 (low-level) evidence

[48]

Prabhakar A, Kaye AD, Wyche MQ, Salinas OJ, Mancuso K, Urman RD. Novel drug development for neuromuscular blockade. Journal of anaesthesiology, clinical pharmacology. 2016 Jul-Sep:32(3):376-8. doi: 10.4103/0970-9185.188833. Epub     [PubMed PMID: 27625489]


[49]

Thilen SR, Weigel WA, Todd MM, Dutton RP, Lien CA, Grant SA, Szokol JW, Eriksson LI, Yaster M, Grant MD, Agarkar M, Marbella AM, Blanck JF, Domino KB. 2023 American Society of Anesthesiologists Practice Guidelines for Monitoring and Antagonism of Neuromuscular Blockade: A Report by the American Society of Anesthesiologists Task Force on Neuromuscular Blockade. Anesthesiology. 2023 Jan 1:138(1):13-41. doi: 10.1097/ALN.0000000000004379. Epub     [PubMed PMID: 36520073]

Level 1 (high-level) evidence

[50]

Messieha ZS, Guirguis S, Hanna S. Bispectral index monitoring (BIS) as a guide for intubation without neuromuscular blockade in office-based pediatric general anesthesia: a retrospective evaluation. Anesthesia progress. 2011 Spring:58(1):3-7. doi: 10.2344/0003-3006-58.1.3. Epub     [PubMed PMID: 21410358]

Level 2 (mid-level) evidence

[51]

Short TG, Campbell D, Leslie K. Response of bispectral index to neuromuscular block in awake volunteers. British journal of anaesthesia. 2016 May:116(5):725-6. doi: 10.1093/bja/aew089. Epub     [PubMed PMID: 27106988]


[52]

Messner M, Beese U, Romstöck J, Dinkel M, Tschaikowsky K. The bispectral index declines during neuromuscular block in fully awake persons. Anesthesia and analgesia. 2003 Aug:97(2):488-491. doi: 10.1213/01.ANE.0000072741.78244.C0. Epub     [PubMed PMID: 12873942]


[53]

Schuller PJ, Newell S, Strickland PA, Barry JJ. Response of bispectral index to neuromuscular block in awake volunteers. British journal of anaesthesia. 2015 Jul:115 Suppl 1():i95-i103. doi: 10.1093/bja/aev072. Epub     [PubMed PMID: 26174308]


[54]

Bom A, Bradley M, Cameron K, Clark JK, Van Egmond J, Feilden H, MacLean EJ, Muir AW, Palin R, Rees DC, Zhang MQ. A novel concept of reversing neuromuscular block: chemical encapsulation of rocuronium bromide by a cyclodextrin-based synthetic host. Angewandte Chemie (International ed. in English). 2002 Jan 18:41(2):266-70     [PubMed PMID: 12491405]

Level 3 (low-level) evidence

[55]

Pühringer FK, Rex C, Sielenkämper AW, Claudius C, Larsen PB, Prins ME, Eikermann M, Khuenl-Brady KS. Reversal of profound, high-dose rocuronium-induced neuromuscular blockade by sugammadex at two different time points: an international, multicenter, randomized, dose-finding, safety assessor-blinded, phase II trial. Anesthesiology. 2008 Aug:109(2):188-97. doi: 10.1097/ALN.0b013e31817f5bc7. Epub     [PubMed PMID: 18648227]

Level 1 (high-level) evidence

[56]

de Boer HD, Driessen JJ, Marcus MA, Kerkkamp H, Heeringa M, Klimek M. Reversal of rocuronium-induced (1.2 mg/kg) profound neuromuscular block by sugammadex: a multicenter, dose-finding and safety study. Anesthesiology. 2007 Aug:107(2):239-44     [PubMed PMID: 17667567]

Level 1 (high-level) evidence

[57]

Groudine SB, Soto R, Lien C, Drover D, Roberts K. A randomized, dose-finding, phase II study of the selective relaxant binding drug, Sugammadex, capable of safely reversing profound rocuronium-induced neuromuscular block. Anesthesia and analgesia. 2007 Mar:104(3):555-62     [PubMed PMID: 17312208]

Level 1 (high-level) evidence

[58]

Blobner M, Frick CG, Stäuble RB, Feussner H, Schaller SJ, Unterbuchner C, Lingg C, Geisler M, Fink H. Neuromuscular blockade improves surgical conditions (NISCO). Surgical endoscopy. 2015 Mar:29(3):627-36. doi: 10.1007/s00464-014-3711-7. Epub 2014 Aug 15     [PubMed PMID: 25125097]

Level 1 (high-level) evidence

[59]

Barrio J, Errando CL, García-Ramón J, Sellés R, San Miguel G, Gallego J. Influence of depth of neuromuscular blockade on surgical conditions during low-pressure pneumoperitoneum laparoscopic cholecystectomy: A randomized blinded study. Journal of clinical anesthesia. 2017 Nov:42():26-30. doi: 10.1016/j.jclinane.2017.08.005. Epub 2017 Aug 30     [PubMed PMID: 28803124]

Level 1 (high-level) evidence

[60]

Martini CH, Boon M, Bevers RF, Aarts LP, Dahan A. Evaluation of surgical conditions during laparoscopic surgery in patients with moderate vs deep neuromuscular block. British journal of anaesthesia. 2014 Mar:112(3):498-505. doi: 10.1093/bja/aet377. Epub 2013 Nov 15     [PubMed PMID: 24240315]

Level 1 (high-level) evidence

[61]

Boon M, Martini CH, Aarts LP, Bevers RF, Dahan A. Effect of variations in depth of neuromuscular blockade on rating of surgical conditions by surgeon and anesthesiologist in patients undergoing laparoscopic renal or prostatic surgery (BLISS trial): study protocol for a randomized controlled trial. Trials. 2013 Mar 1:14():63. doi: 10.1186/1745-6215-14-63. Epub 2013 Mar 1     [PubMed PMID: 23452344]

Level 1 (high-level) evidence

[62]

Kollef MH. Evaluating the Value of the Respiratory Therapist: Where Is the Evidence? Focus on the Barnes-Jewish Hospital Experience. Respiratory care. 2017 Dec:62(12):1602-1610. doi: 10.4187/respcare.05807. Epub     [PubMed PMID: 29162728]