Neuromuscular Disorders Including Malignant Hyperthermia and Other Genetic Disorders





Key Words

Charcot-Marie-Tooth Disease, Dantrolene, Guillain-Barre Syndrome, Malignant Hyperthermia, Multiple Sclerosis, Muscular Dystrophy, Ryanodine

 




Key Points





  • Malignant hyperthermia (MH) is a pharmacogenetic disorder inherited primarily in an autosomal dominant pattern.



  • MH susceptibility is linked to 230 mutations in the skeletal muscle ryanodine receptor (RyR1) and four mutations in the calcium voltage-gated channel subunit alpha1 S (CACNA1S) genes that encode two Ca 2+ channels necessary for skeletal muscle excitation-contraction coupling.



  • Physical interactions between L-type Ca 2+ channel (Ca v 1.1) and RyR1 tightly regulate initiation and termination of skeletal muscle excitation-contraction coupling.



  • Skeletal muscle accounts for approximately 40% of body weight and inherent changes in its metabolism have profound impacts on whole-body metabolism and physiology.



  • Carriers of MH mutations can exhibit mild to moderate muscle impairments in the absence of triggering agents but are rarely diagnosed.



  • Carriers of MH mutations are susceptible to anesthetic-triggered runaway skeletal muscle metabolism, which if not promptly treated is lethal.



  • S igns of MH, including increased end-tidal CO 2 , increased core temperature, muscle rigidity, tachycardia, and more , are consequences of the fulminant hypermetabolic crisis.



  • Exposure to triggering agents or heat stress leads to acute loss of RyR1/Ca v 1.1 channel regulation, rapid accumulation of Ca 2+ within the sarcoplasm, and a hypermetabolic crisis that stimulates adenosine triphosphate (ATP) utilization by pumps attempting to restore resting Ca 2+ balance among sarcoplasmic reticulum, mitochondrial, and extracellular compartments.



  • Dantrolene markedly attenuates myoplasmic calcium (Ca 2+ ) concentrations and thereby restores resting Ca 2+ balance and metabolism, with reversal of clinical signs.



  • Evaluation of persons susceptible to MH includes an in vitro contracture test (IVCT) and caffeine/halothane contracture test (CHCT), and evaluation of DNA to identify mutations.



  • Currently DNA testing alone can be used to evaluate 42 human mutations and all swine, equine, and canine MH.



  • Future MH goals include advancement of genetic evaluations in North American and European medical programs and stronger finances to support genetic studies, the identification of the mode of action of dantrolene, a determination of the immediate cause of triggering MH, and the development of effective, noninvasive tests for MH susceptibility.



  • The absence of mutations in dystrophin, along with dystrophin-associated glycoproteins, is involved in sarcolemmal stability. Its defects are responsible for Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD).



  • Whereas the risk for an MH mutation in DMD and BMD patients is similar to that in the general population, the incidence of MH-like anesthetic events has been reported to be 0.002 with DMD and 0.00036 with BMD.



  • Succinylcholine is contraindicated in DMD and BMD patients because of the risk of rhabdomyolysis and hyperkalemia as a result of their unstable sarcolemmal membrane.



  • Reversal of neuromuscular blockade with sugammadex is a practical alternative to the management of many of these disorders, if rocuronium or vecuronium is used. The combination of rocuronium and sugammadex has improved the anesthetic management for some of these challenging disorders.





Acknowledgment


This chapter is a consolidation of two chapters in the 8th edition, Chapter 42 , Neuromuscular Disorders and other Genetic Disorders and Chapter 43 , Malignant Hyperthermia and Muscle-Related Disorders. The editors, publisher, and the returning authors, would like to thank the following authors: Aranya Bagchi, Richa Saxena, and Diptiman Bose for their contributions to the prior edition of this work. It has served as the foundation for the current.




Malignant Hyperthermia


Malignant hyperthermia (MH) is one of the most devastating anesthesia-related complications. The fulminant MH syndrome is elicited by the administration of triggering anesthetic agents, such as volatile halogenated anesthetics or depolarizing neuromuscular blocking agents (NMBAs). MH has been and continues to be a life-threatening complication of anesthesia if the diagnosis is not made promptly and treatment is not begun in a timely fashion. Unlike other disorders described in this chapter, MH has virtually no characteristic phenotype before exposure to the triggering agent and is truly an example of the interaction of genes and the environment. Also covered in this chapter are some of the neuromuscular disorders, although rarely encountered in a routine anesthetic practice. This group of disorders challenges both perioperative management and intensive care. They affect the normal function of peripheral nerves, the neuromuscular junction, and/or muscles. Although such diseases are thought to be rare, the number of patients that a clinician may encounter is increasing because of better medical care, increasing longevity, and other possible unidentified factors. Neuromuscular disorders have significant potential to interact with an improper anesthetic plan, and all affected patients require special perioperative attention for anesthetic management. In this area, the armamentarium of invasive and noninvasive diagnostic tools is being developed, especially in genetics.


MH is a pharmacogenetic clinical syndrome that, in its classic form, occurs during anesthesia with volatile halogenated alkanes such as halothane, isoflurane/sevoflurane, /desflurane, and/or administration of the depolarizing muscle relaxant succinylcholine. The fulminant MH episode observed clinically produces muscle hypermetabolism with rapidly increasing body temperature, by as much as 1°C in 5 minutes, and extreme acidosis as a result of acute loss of control of intracellular ionized calcium (Ca 2+ ). It is the sustained high levels of sarcoplasmic Ca 2+ that rapidly drives skeletal muscle into a hypermetabolic state that may proceed to severe rhabdomyolysis. Although MH was initially associated with a mortality rate of 60%, earlier diagnosis and the use of dantrolene have reduced the mortality to less than 1.4%. Current cases of MH are restricted in severity because of diagnostic awareness, early detection through end-expired carbon dioxide (CO 2 ), the use of less potent anesthetic triggers, and prior administration of drugs that attenuate the progression of the fulminant episode. Estimates of the incidence of fulminant MH vary widely from one case per 10,000 to 1:250,000 anesthetics administered. The prevalence of MH events in Japan was calculated to be between 1:60,000 and 1:73,000. However, the prevalence of MH mutations within kindred known to transmit MH-susceptibility (MHS) mutations may be as high as 1:2000. Males appear to be more susceptible to developing a clinical MH episode than females. A gender difference in MHS has also been demonstrated in knock-in mice expressing human MH mutation RyR1-T4825I. The pediatric population accounts for 52.1% of all MH reactions.


Between 50% and 80% of genotyped patients who have had a clinical MH syndrome and a positive muscle biopsy have had their disease linked to one of more than 230 mutations in the type 1 ryanodine receptor (RyR1; sarcoplasmic reticulum [SR] Ca 2+ release channel) gene and four mutations in L-type Ca 2+ channel (Ca V 1.1), the pore subunit of the slowly inactivating L-type Ca 2+ channel encoded by Calcium Voltage-Gated Channel Subunit Alpha1 S (CACNA1S) (also referred to as the dihydropyridine receptor [DHPR]). The genetics of MHS and the related abnormal function of RyR1, the DHPR, and associated proteins are being investigated at the molecular biologic level, with a porcine model and several new mouse models providing intricate details about the etiology of the disorder. Parallel studies in humans are limited by scarce material for scientific study and are complicated by the fact that phenotypes within a single genotype vary as a result of sex, age, genetic, epigenetic, and environmental modifiers.


Public education and communication in the United States are provided by Malignant Hyperthermia Association of the United States (MHAUS, 11 E. State Street, P.O. Box 1069, Sherburne, NY 13460, U.S.A.; telephone: (+1) 607-674-7901; fax: (+1) 607-674-7910; e-mail: info@mhaus.org ; website: http://www.mhaus.org ), and by emergency consultation with the MH Hotline (1-800-MHHYPER, or 1-800-644-9737). The North American Malignant Hyperthermia Registry (NAMHR), a professional subsidiary of MHAUS, collates findings from muscle biopsy centers in Canada and the United States (NAMHR, 1345 SW Center Drive, P.O. Box 100254, Gainesville, FL 32610, U.S.A.); telephone: (+1) 888-274-7899; fax: (+1) 352-392-7029.; website http://anest.ufl.edu/namhr/ ).


History


Between 1915 and 1925, one family experienced three anesthetic-induced MH deaths with rigidity and hyperthermia and was puzzled for decades regarding the cause of these deaths. MHS was eventually confirmed in three descendants by in vitro muscle biopsy tests. In 1929, Ombrédanne described anesthesia-induced postoperative hyperthermia and pallor in children accompanied by significant mortality but did not detect any familial relationships. Critical worldwide attention to MH began in 1960 when Denborough and associates reported a 21-year-old Australian with an open leg fracture who was more anxious about anesthesia than about surgery because 10 of his relatives died during or after anesthesia. Denborough and colleagues initially anesthetized him with the then-new agent halothane, halted it when signs of MH appeared, successfully treated the symptoms, aborted the syndrome, and subsequently used spinal anesthesia. Further evaluations by George Locher in Wausau, Wisconsin, and Beverly Britt in Toronto, Canada, led to the discovery that MH risk was indeed familial. It was also found that the cause of the syndrome was the result of skeletal muscle involvement rather than central loss of temperature control by the recognition of increased muscle metabolism or muscle rigidity early in the syndrome, low-threshold contracture responses, and elevated creatine kinase (CK) values.


Interestingly, a similar syndrome was discovered in swine inbred with breeding patterns designed to produce a rapid growth rate and superior muscle development (e.g. , Landrace, Piétrain, Duroc, and Poland China). Porcine stress syndrome , which is associated with increased metabolism, acidosis, rigidity, fever, and death from rapid deterioration of muscle and results in pale, soft, exudative pork, can be triggered by any stress, such as separation, shipping conditions, weaning, fighting, coitus, or preparation for slaughter, and had become a significant problem for meat production. In 1966, Hall and coworkers reported that a syndrome that appeared to be identical to MH could be induced in stress-susceptible swine by the administration of halothane and succinylcholine. The cause of this syndrome in pigs was discovered to be a single missense mutation in RyR1 , and all susceptible swine have the same Arg615Cys mutation in the SR calcium release channel RyR1.


In 1975, Harrison described the efficacy of dantrolene in preventing and treating porcine MH, which was rapidly confirmed in humans by a multihospital evaluation of dantrolene used to treat anesthetic-induced MH episodes. Today, dantrolene still remains the primary pharmacologic approach for successful MH therapy.


Physiology and Pathophysiology of Excitation-Contraction Coupling and Malignant Hyperthermia


MH is a syndrome caused by dysregulation of excitation-contraction (EC) coupling in skeletal muscle. Normal muscle contraction is initiated by nerve impulses arriving at the neuromuscular junction (i.e. , the motor end plate) that trigger the release of acetylcholine from the nerve terminal. Acetylcholine activates nicotinic cholinergic receptors (nAChR), nonselective cation channels located at the postsynaptic neuromuscular junction, that are essential for local depolarization of the surface muscle membrane (sarcolemma) and initiating action potentials that propagate rapidly along the sarcolemma of muscle cells. Invaginations of the sarcolemma (termed transverse or T tubules) act as conduits to rapidly and uniformly direct-action potentials deep within the myofibrils where they transduce a conformational change in the “voltage sensor” protein Ca V 1.1. A central T-tubule is flanked on both sides by a terminal cisternae element from the SR that contains the Ca 2+ release channels (RyR1). Conformational changes in Ca V 1.1 residing within the T-tubule are mechanically transmitted to RyR1 residing in the junctional face of the SR. More specifically, physical coupling of four Ca V 1.1 (dihydropyridine receptor) units to every second RyR1 channel form linear arrays at specialized junctions ( triadic junctions ) that are essential for linking electrical signals at the T tubules with the release of Ca 2+ stored within the SR. Release of SR Ca 2+ causes the free, cytoplasmic (sarcoplasmic) Ca 2+ concentration to increase from 10 –7 M to about 10 –5 M. This released Ca 2+ binds to contractile proteins (troponin C and tropomyosin) in the thin filament to expose myosin’s actin binding sites which allow shortening and force development by the muscle fibers (i.e. , muscle contraction). The entire process is termed excitation-contraction coupling (EC coupling) Intracellular Ca 2+ pumps (i.e. , sarcoplasmic/endoplasmic reticulum Ca 2+ -adenosine triphosphatase [ATPase], or SERCA) rapidly sequester Ca 2+ back into the SR lumen, and muscle relaxation begins when the Ca 2+ concentration falls below 10 –6 M and ends when the resting sarcoplasmic Ca 2+ concentration is restored to 10 –7 M. Because both contraction and relaxation are energy-related processes that consume adenosine triphosphate (ATP), knowing the molecular events contributing to EC coupling and the subsequent relaxation phase is essential to understanding the cause of MH ( Fig. 35.1 ). Clinical and laboratory data from humans, swine, and mice with knock-in mutations indicate that the fulminant MH syndrome is associated with a persistent increase in the concentration of sarcoplasmic Ca 2+ . The increased activity of pumps and exchangers trying to correct the increase in sarcoplasmic Ca 2+ associated with triggered MH increases the need for ATP, which in turn produces heat. Thus the common etiological feature of the disorder is hyperthermia. The rigidity that is frequently seen during a fulminant MH episode is the result of the inability of the Ca 2+ pumps and transporters to reduce the unbound sarcoplasmic Ca 2+ below the contractile threshold (10 –6 M). Dantrolene is an effective therapeutic for treatment of fulminant MH because it reduces the concentration of sarcoplasmic Ca 2+ to below contractile threshold. However, the pathway by which dantrolene lowers sarcoplasmic Ca 2+ is complex and still not fully understood. Dantrolene’s ability to suppress Ca 2+ release from SR appears to depend on elevated sarcoplasmic Mg 2+ concentration ; however the drug also attenuates depolarized-triggered Ca 2+ entry mediated by Ca V 1.1, which is exacerbated in MHS muscle cells and MH normal muscle cells exposed to ryanodine. Thus whether dantrolene directly inhibits RyR1 or requires additional intermediates within the triad junctions remains to be clarified.




Fig. 35.1


Key ion channels involved in neuromuscular transmission and excitation-contraction coupling. Nerve impulses arriving at the nerve terminal activate voltage-gated Ca 2+ channels (1). The resulting increase in cytoplasmic Ca 2+ concentration is essential for the exocytosis of acetylcholine. Binding of acetylcholine to postsynaptic nicotinic cholinergic receptors (nAChR) activates an integral nonselective cation channel that depolarizes the sarcolemma (2). Depolarizing the sarcolemma to threshold activates voltage-gated Na + channels (3), which initiates action potential impulses that propagate deep into the muscle through the transverse tubule system. Within the transverse tubule system, L-type voltage-gated Ca 2+ channels sense membrane depolarization and undergo a conformational change (4). A physical link between these voltage sensors and the ryanodine receptor (RyR1) sarcoplasmic reticulum Ca 2+ channel is the means by which the electrical signal is transferred from the T tubule to Ca 2+ release from the sarcoplasmic reticulum (5).

Modified from Alberts B, Bray D, Lewis J, et al. Molecular Biology of the Cell . 3rd ed. New York: Garland Press; 1994.




Malignant Hyperthermia Is the Result of Abnormal Function of Muscle Calcium Release Units


Ryanodine Receptors


Ryanodine receptors (RyRs) within the muscle are synonymous with the junctional foot protein/SR calcium release channel, and are so named because they specifically bind the toxic plant alkaloid ryanodine, which can activate or inhibit the channel depending on its concentration. In all mammals there are three RyR isoforms. In humans, they are encoded by three genes located on chromosomes 19q13.1, 1q42.1-q43, and 15q14-q15, for the “skeletal” (RyR1), “cardiac” (RyR2), and “brain” (RyR3) isoforms, respectively. Each functional RyR is a homotetramer consisting of four identical subunits (∼5000 amino acids each), and an accessory protein, calstabin 1 (FK506 12-kd binding protein [FKBP12]). . The total mass of the tetramer exceeds 2 mega-Daltons. Thus it is one of the largest known proteins and the largest known channel in mammalian species. Evidence of direct coupling of Ca V 1.1 and RyR1 has been demonstrated both by expressing chimeric Ca V 1.1/Ca V 1.2 cDNA in dysgenic myotubes that lack constitutive expression of Ca V 1.1 and chimeric RyR1/RyR2/3 cDNA in dyspedic myotubes that lack constitutive expression of RyR1, 2, and 3. Such studies have provided compelling evidence that the cytoplasmic region between repeats II and III (i.e. , cytosolic II-III loop) of Ca V 1.1 contains a stretch of 46 amino acids (L720 to Q765) and multiple regions of RyR1 that are essential for engaging bidirectional signaling between Ca V 1.1 and RyR1.


In the last two decades, our understanding of EC coupling has increased significantly by identifying protein-protein interactions that regulate both the release and sequestration of Ca 2+ within skeletal muscle. The elemental unit of function has been named the Ca 2+ release unit (CRU), and it is localized within junctional regions of T-tubule and SR membranes. The CRU is a macromolecular assembly of interacting proteins that participate in regulating EC coupling. RyR1 is a high-conductance channel that regulates release of SR Ca 2+ and is the central component of the CRU. The functional RyR1 tetramer anchored within the SR membrane physically spans the junctional space to interact with tetrads composed of four voltage-activated Ca V 1.1 subunits within the T-tubule membrane. This physical interaction engages a form of bidirectional signaling that tightly regulates the function of both proteins. Moreover, interaction of Ca V 1.1 and RyR1 does not occur in isolation, but are further subject to regulation by a number of proteins localized within the triad junction, including Homer 1, which physically binds and functionally couples target proteins, calstabin 1, triadin, junctin, Mg29, junctophilin 1 and 2, calsequestrin, calmodulin, STAC 3, the catalytic and regulatory subunits of protein kinase A, and protein phosphatase 1. . It is likely that this list is not complete and that there are other critical components which make up this tightly regulated macromolecular complex. More importantly, there is increasing experimental evidence that mutations found in RyR1 ( MH RyR) or Ca V 1.1 ( MH Ca V 1.1) can alter protein-protein interactions in the CRU, as well as alter the functional fidelity of bidirectional signals.


In the presence of certain chemical substances, MH mutations in RyR1 or DHPR cause severe dysregulation of RyR1 channel function. This can be seen in vitro as a heightened sensitivity to volatile anesthetics, 4-chloro- m -cresol, caffeine, ryanodine, and potassium depolarization. Chemically induced dysfunction of the RyR1 complex appears to be the principal cause of triggering uncontrolled skeletal muscle metabolic acidosis (aerobic and glycolytic), rigidity, and hyperkalemia, but the mechanisms governing the syndrome are unclear. Also unclear is the relationship among exertional heat illness, exertional rhabdomyolysis, and MHS, an area that requires more investigation and, if possible, controlled clinical studies.


Two essential cations greatly shape the kinetics and magnitude of Ca 2+ release in response to depolarizing triggers: Ca 2+ itself and Mg 2+ . The normal RyR1 complex responds to Ca 2+ in a biphasic manner. First, Ca 2+ activates the channel in a graded manner between 100 nM and 100 μM, whereas higher concentrations inhibit channel activity. This biphasic action is thought to occur via binding of Ca 2+ to two classes of regulatory sites on RyR1, a high-affinity stimulatory site and a low-affinity inhibitory site. Mg 2+ -induced inhibition is the second important physiologic regulator of RyR1 activity in skeletal muscle. Mg 2+ inhibits RyR1 in a cooperative manner (n H ≈ 2; 50% inhibitory concentration [IC 50 ] ≈ 650 μM). It is likely that Mg 2+ acts by competing with Ca 2+ at its activator sites and by binding to yet unidentified low-affinity inhibitory sites. It is possible that MH mutations introduce allosteric instability into the RyR1 complex which leads to a reduction of inhibition rather than directly altering the binding properties of Ca 2+ or Mg 2+ , or both, at the activator or inhibitor sites. Therefore hypersensitivity to pharmacologic agents is likely to be closely tied to altered responses to physiologic ligands. However, whether MHS channels are primarily hyposensitive to inhibition by Mg 2+ or Ca 2+ (or both), are hypersensitive to activation by Ca 2+ , or exhibit altered sensitivities in both directions to both ions seems to be highly dependent on the location of the MH mutation. Studies have also pursued the “leaky channel” hypothesis by examining SR preparations from homozygous R615C MHS pigs and heterozygous R163C and C512S mice. They observed a significantly lower Ca 2+ loading capacity (38%, 23%, and 22% lower than matched wild type mice, respectively) primarily mediated by the presence of leaky channels that remain active even with 100 nM extravesicular Ca 2+ . Recent studies indicated that expression of Ca V 1.1 represses the basal activity of the ryanodine-insensitive RyR1 leaky state. It is important that MHS mutations appear to not only alter bidirectional signaling during EC coupling and inherent regulation of RyR1 channel functions, but also weaken negative regulation conferred by Ca V 1.1 on RyR1 Ca 2+ leak under nontriggering conditions. These findings at the molecular and cellular level using knock-in MHS mice confirm earlier measurements made in porcine and human MHS muscles, myotubes, and myoball preparations and in dyspedic myotubes expressing MH RyR1 cDNAs, all of which have been shown to have chronically elevated resting cytoplasmic [Ca 2+ ] i .


Results from both functional and structural evidence suggests that long-range interdomain interactions between regions of RyR1 are involved in channel regulation by stabilizing protein conformations critical for normal channel transitions. A three-dimensional reconstruction of RyR1 by Samso and coworkers shows that the RyR1 architecture is designed to support long-range allosteric pathways such as coupling with Ca V 1.1 and binding to ligands such as calmodulin and FKBP12. This structural model for gating has been recently confirmed at molecular scale resolution by several laboratories.


Voltage-Gated Calcium Channels: role of Ca v 1.1


Although the majority of mutations that confer MHS reside in the RyR1 gene, three mutations in the CACNA1S gene encoding for the Ca V 1.1 subunit of skeletal muscle have been linked to human MHS. The Arg1086His mutation in the intracellular loop connecting homologous repeats III and IV of Ca V 1.1 represented the first MH-causing mutation so far identified in a protein other than RyR1. Physiological characterization of the R1086H mutation further demonstrated that sensitivity of RyR1 activity was significantly enhanced by membrane depolarization or by pharmacologic activators of RyR1 (e.g. , caffeine). In addition, Pirone and associates have identified an MHS-causing Thr1354Ser mutation in the S5-S6 extracellular pore-loop region of the homologous repeat IV of Ca V 1.1 Expression of the T1354S mutation also accelerated L-type Ca 2+ current kinetics and also contributed to an increase in RyR1-mediated Ca 2+ release. The Arg174Trp Ca V 1.1 MH mutation occurs at the innermost basic residue of the IS4 voltage-sensing helix, a residue conserved among all Ca V channels. Unlike the other Ca V 1.1 MHS mutations, homozygous expression of R174W completely ablates the L-type current, but despite this, has no influence on normal EC coupling. In murine studies, muscle fibers from Het R174W animals verify the increased sensitivity of Ca 2+ release to caffeine and halothane compared with myotubes expressing wild type Ca V 1.1, but whether this mutation is sufficient to confer anesthetic- or heat-triggered fulminant MH remains to be tested.


Factors Other than Ryanodine Receptor Abnormalities


Other cellular processes can affect MH episodes. It has been demonstrated that concurrent administration of nondepolarizing neuromuscular blocking drugs at the same time as triggering agents can delay or prevent the onset of clinical MH syndrome. Pretreatment of MHS pigs with sufficient nondepolarizing neuromuscular blocking agent, which is used to completely abolish muscle twitch elicited by electrical stimulation of the nerve, prevented halothane from triggering the clinical syndrome for 90 minutes, the longest time point tested. However, in the continued presence of halothane, when function of the neuromuscular junction was restored by administration of the cholinesterase inhibitor neostigmine, clinical MH was triggered immediately. This suggested a close relationship between functional neuromuscular junctions or depolarization of the sarcolemma (or both) and the clinical syndrome.


In myotubes, sarcolemmal excitation-coupled Ca 2+ entry (ECCE) is sensitive to the conformation of the RyR1 and is enhanced by several mutations in RyR1, including MH mutations. ECCE appears to be an inherent property of Ca V 1.1 during long or repetitive depolarization of myotubes, possibly mediated by shifting Ca V 1.1 to the mode 2 gating conformation. Nevertheless, enhanced ECCE in MHS muscle may contribute to an increased sensitivity to depolarization and appears to be one target for dantrolene’s abrogation of responses to both electrical and potassium chloride depolarization. Although CACNA1S expression undergoes developmental switching to a splice variant that downregulates Ca 2+ current density of Ca V 1.1 channels in adult fibers, mutations that maintain Ca 2+ current density more similar to those measured with embryonic myotubes has recently been shown to promote muscle pathology.


In addition to ECCE, classic store-operated capacitive Ca 2+ entry pathways similar to the store-operated Ca 2+ entry (SOCE) seen in nonexcitable cells have been shown to be present in skeletal muscle and appear to be more active in MHS muscles both at rest as a response to chronic store depletion and during an MH crisis. These SOCE channels have also been suggested to be a target for dantrolene, but this has not been validated by other studies. Together, these data suggest that MH RyRs or MH Ca V 1.1 assume a conformation that enhances Ca 2+ entry via ECCE or SOCE (or both). This enhanced entry, when combined with decreased sensitivity of MH RyRs to Ca 2+ and Mg 2+ inhibition, could provide cellular conditions that heighten sensitivity to triggering agents and perpetuate the fulminant clinical MH syndrome.


Dantrolene


Dantrolene is the only medication that has been shown to be effective in reversing the symptoms of MH. Preadministration of dantrolene will also prevent the development of fulminant MH in homozygous pigs or MH mice when exposed to a triggering stimulus. Dantrolene sodium is a hydantoin derivative (1-[5-(4-nitrophenyl)-2-furanyl]methylene]imino]-2,4-imidazolidinedione) that does not block neuromuscular transmission, but causes muscle weakness by direct muscular action. The properties of dantrolene have been closely correlated with its ability to reduce efflux of Ca 2+ from the SR in vitro. Dantrolene (20 μM) counteracts the effect of reduced Mg 2+ inhibition in MH-affected muscle. Dantrolene (20 μM) can inhibit the enhanced sensitivity to caffeine seen in MH muscles, and both dantrolene and its more water-soluble analog azumolene (150 μM) have been shown to reduce depolarization-induced release of Ca 2+ , both in muscle and in triadic vesicles. The idea that dantrolene suppresses SR Ca 2+ release as a result of direct interactions with RyR1 is somewhat controversial. Paul-Pletzer and associates demonstrated that [ 3 H]azidodantrolene specifically labels the amino terminus of RyR1 defined by the 1400-amino acid residue N-terminal calpain digestion fragment of RyR1. More detailed analysis further localized the [ 3 H]azidodantrolene binding site to a single domain containing the core sequence corresponding to amino acid residues 590 through 609 of RyR1. However, to date, we lack evidence of a direct action of dantrolene on single RyR1 channels studied in lipid bilayers, even though they are reconstituted with calstabin 1, ATP, and activating concentrations of Ca 2+ , which suggests that dantrolene’s main action is to alter key protein-protein interactions. The recent discovery that inhibition of SR Ca 2+ release by dantrolene requires Mg 2+ may help resolve the controversy of the conflicting observations on dantrolene inhibition of RyR1 channel activity in Mg 2+ -free bilayer experiments.




Genetics


RyR1 mutations have been found in 50% to 80% of patients and relatives who are labeled MHS by positive contracture tests and in almost all families with central core disease (CCD) and King-Denborough syndrome (KDS). More than 210 missense mutations and 8 deletions associated with MH have thus far been detected. Another 29 missense mutations are associated with CCD and multiminicore disease ( MmD ) in patients with unknown MH testing status. Interestingly, 40% of missense RyR1 mutations occur at CpG dinucleotide sequences. Five other loci (17q21-24, 1q32, 3q13, 7q21-24, and 5p) have been linked to families with both positive contracture tests and an unusual response to anesthesia, and have been designated MHS loci 2 through 6, respectively. However, of these five, the only gene that has been shown to be associated with MH is CACNA1S , which codes for Ca V 1.1 (the α 1S -subunit of DHPR) in the MHS3 locus. Two causative mutations in this gene are linked to less than 1% of MHS families worldwide. In some of the other loci, all genes within the locus have been ruled out as causing susceptibility to MH. Hence for practical reasons, the RyR1 gene remains the primary target for current clinical genetic analysis.


Distribution of RyR1 Mutations


The missense mutations associated with MHS, CCD, or, in some cases, both, are dispersed throughout the coding region of the RyR1 gene, and all allow transcription of a protein that is putatively functional. Until recently, it was thought that most RyR1 mutations were clustered in three “hot spots:” between amino acid residues 35 and 614 (MH/CCD region 1), between amino acid residues 2163 and 2458 (MH/CCD region 2) in the sarcoplasmic foot region of the protein, and between amino acids 4643 and 4898 in the carboxyl-terminal transmembrane loop or pore region (MHS/CCD region 3) ( Fig. 35.2 ). It appears that the supposition that there were “hot spots” was simply due to bias in sample analysis inasmuch as the missense mutations associated with MH or CCD (or both) are scattered over 54 of the 107 exons of RyR1 . Approximately 41% of reported MH mutations are found in multiple families. CCD mutations are predominantly found in the C-terminal region of the gene (exons 85-103), and only 10 mutations (17%) have been described in more than one family: R4861H ( n = 14), V4849I ( n = 9), I4898T ( n = 7), L4824P ( n = 4), A4940T ( n = 4), G4638D ( n = 3), R4893W ( n = 3), R4861C ( n = 2), R4893Q ( n = 2), and G4899E ( n = 2).




Fig. 35.2


Schematic representation of the triad junction of skeletal muscle shows the junctional foot protein (ryanodine [RyR1] receptor) and its associated proteins. In skeletal muscle, the α 1S -subunit of the dihydropyridine receptor (DHPR) participates in excitation-contraction coupling. These physical links transmit essential signals across the narrow gap of the triadic junction that activate the RyR1 and release Ca 2+ from the sarcoplasmic reticulum.

Modified from Pessah IN, Lynch C III, Gronert GA: Complex pharmacology of malignant hyperthermia. Anesthesiology 1996;84:1275.


The true ethnic distribution of MH and CCD is difficult to ascertain. MH and CCD have been reported in Western populations predominantly, but this is likely the result of the manner and frequency in which cases are reported. It does appear that some mutations are clustered in a given region of the world, but the distribution and frequency appear to be somewhat population specific. In the United Kingdom, 69 RyR1 mutations have been discovered, 25 of which are found only in a single family. G2434R is found in approximately 40% of the 434 mutation-positive MH families investigated in the United Kingdom, with the next most common mutations being T2206M (10%) and G341R (8%). In Switzerland, V2168M and I2336H are the predominant mutations, and in Germany, R163C (MH and CCD), R614C (MH), T2206M (MH), G2434R (MH), and R2454H (MH) have each been detected in five or more independent cases. G341R and R614C are common in France, and R614C has also been found in several MH families from Italy and Canada. G341R has been found frequently in Belgium. The mutation common to Europe and North America is G2434R, which occurs in 4% to 7% of European and 5.5% of North American families. Single-family mutations are the most common mutations reported in Japanese, Chinese, Taiwanese, Australian, and New Zealand MH families with the exception of the R163C mutation reported in a large population in rural New South Wales in Australia and the T4826I mutation which is found in numerous families in the Maori population of New Zealand. However, despite these two exceptions, it is likely that the reason for unique family mutations in Asia and Australasia may reflect the small number of cases investigated there. Because genetic screening in European and North American studies has predominantly targeted only regions 1 and 2 of the original hot spots in the gene, the absence of RyR1 mutations in some of the screened population could be explained by RyR1 mutations located outside these two regions or by involvement of other genes.


Inheritance and Penetrance of Malignant Hyperthermia


Inheritance of human MH can no longer be considered to be solely autosomal dominant with variable penetrance because more than one MH-linked mutation has been identified in some probands and families. Six non-consanguineous families harbor at least two RyR1 mutations that have both been linked to MHS, and in two of those families, one is an RyR1 mutation and the second is a Ca V 1.1 mutation. Although MHS homozygotes are common in affected pigs, they are rare in humans and only found in 50% of currently available transgenic mouse populations. The known MHS homozygous humans also appear clinically normal, but exhibit stronger responses to in vitro contracture test (IVCT) and caffeine/halothane contracture tests (CHCTs) than heterozygous individuals do. Homozygosity of two MH mutations in “hot spot” 1 leads to perinatal lethality in mice. Double heterozygous individuals do not appear to show any additive effect of the second mutation on IVCT.


In Vitro Contracture Test and Caffeine Halothane Contracture Test


The gold standard for diagnosis of MH is the halothane and caffeine muscle contracture test, also known as the IVCT or the CHCT. There are two protocols developed by the European Malignant Hyperthermia Group (EMHG) and the North American Malignant Hyperthermia Group (NAMHG), respectively. The two protocols are similar, but not identical. For the purpose of differentiation, we designate IVCT to the EMHG protocol and CHCT to the NAMHG protocol.


For IVCT, the muscle biopsy will be performed on the quadriceps (either vastus medialis or vastus lateralis) and consists of three parts: a static caffeine test, a static halothane test, and a dynamic halothane test. For the static caffeine test, a stepwise increased concentration (0.5, 1, 1.5, 2, 3, 4, and 32 mmol/L) is applied. The lowest concentration of caffeine which produces a sustained increase of at least 0.2 g in baseline tension is reported as the caffeine threshold. Then, the halothane threshold is obtained using the same method by exposing the muscle to halothane concentrations of 0.5%, 1%, 2%, and 3% v/v. The dynamic halothane test is performed with the muscle stretched at a constant rate of 4 mm/min to achieve a force of approximately 3 g and held at the new length for 1 min for a 3 min exposure to halothane. For each cycle, halothane concentration will be increased from 0.5%, 1%, 2%, to 3% v/v: volume (solute) per volume (solvent). The concentration of halothane which produces a sustained increase of at least 0.2 g in the muscle tension compared to the pre-halothane control is defined as the dynamic halothane threshold. The IVCT protocol classifies the patients into three groups: MH Susceptible (MHS HC ) group with a caffeine threshold at the caffeine concentration of 2 mmol or less and a halothane threshold of 2% v/v halothane or less; MH Normal (MHN) group with a caffeine threshold at the caffeine concentration of 3 mmol or more without a halothane response at 2% v/v; MHS H and MHS C groups which describe individuals that used to be classified as MHE (equivocal) who are only responsive to either halothane or caffeine alone. The MHE descriptor was dropped because use of the ‘equivocal’ label outside of its laboratory context has the potential to confuse patients and clinicians unfamiliar with its derivation.


For CHCT, a muscle biopsy can be taken from the following sites in the order of preference: (1) the vastus group, (2) the rectus abdominis, and (3) other muscle groups under special circumstances. Required tests include exposure of muscle to 3% v/v halothane alone and to incremental caffeine concentrations (0.5, 1, 2, 4 mmol, and 8.0 mmol if the response at 4 mmol is <1 g, and 32 mmol) alone. Optional tests include exposure of muscle to a combination of both 1% halothane and incremental caffeine concentrations, and to 2% v/v halothane alone. According to CHCT protocol, an individual is MHS when either of the halothane or the caffeine test is positive, and MHN when both tests are negative.


The sensitivity of IVCT was reported to be 99.0% (95% confidence interval [CI] 94.8%-100%) if the MHE group is considered susceptible and the specificity of IVCT was 93.6% (95% CI 89.2%-96.5%), while the sensitivity and specificity of CHCT was 97% (95% CI 84%-100%) and 78% (95% CI 69%-85%), respectively. Recently, fluoroquinolones and statins, 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, were found to induce significant contractures in MHS muscle bundles, but not in MHN. Ondansetron and 3,4-methylenedioxymethamphetamine (MDMA) may also dose-dependently induce contracture or increase the sensitivity of the contractile apparatus to calcium in both MHS and MHN fibers. Modifications to the IVCT protocol by adding ryanodine or 4-chloro- m -cresol, a RyR-specific agonist, has been reported, but has not been included in the standard protocol. Furthermore, Metterlein and associates studied the possibility of replacing halothane with newer volatile agents in IVCT. At increasing concentration, except for sevoflurane, all newer volatile agents, including enflurane, isoflurane, and desflurane, induced significantly greater contractures in MHS muscle compared to MHN bundles. However, within the MHS muscle bundles, halothane produced significantly higher contractures, and was considered the strongest discriminator for MH using the IVCT protocol. A direct application of high sevoflurane concentration of 8%, instead of the stepwise application, has been shown to induce significantly stronger contractures in MHS subjects. Nevertheless, from the retrospective analysis of the Japanese MH database, there was no evidence between the severity of MH triggered by sevoflurane and isoflurane, or other agents, suggesting sevoflurane is a weak or weaker MH triggering agent.


Discordance Between Genetic and in Vitro Contracture Test/Caffeine/Halothane Contracture Tests Malignant Hyperthermia Testing


Discordance has confounded linkage analysis worldwide. Examples include MHN patients carrying an RyR1 mutation associated with MH and MHS patients who do not carry the familial RyR1 mutation. Several explanations are possible, the most likely being that IVCT/CHCT is not clinically precise and that the thresholds for IVCT or CHCT are inexact. This would lead to errors in determining whether a patient was MHN or MHS. A second possibility is variable penetrance with possible allelic silencing, and a third is that individuals with discordance have mutations in other unknown genes or modifier genes that affect the function of RyR1 and its phenotypic penetrance. The discrepancy between the incidence estimates of MH events and the prevalence estimates also points to possible epigenetic factors at work. Carpenter and associates have suggested that the severity of MHS may be related to the RyR1 variant and mutation found within the highly conserved regions of RyR1 gene. The rarity of large kindreds with MH makes linkage analysis and understanding variability in clinical manifestations difficult. Robinson and associates demonstrated by the transmission disequilibrium test that loci on chromosomes 5 and 7 and, to a lesser extent, loci on chromosomes 1 and 7 influence susceptibility to MH.


Guidelines for Genetic Screening


In 2000, the European MH group (EMHG) formulated guidelines for RyR1 mutation screening with linkage data to other loci for some MH families, but all MH investigators emphasized the vital role of IVCT in the diagnosis of MH. These guidelines for screening have reduced the number of relatives requiring contracture testing without increasing the risk of misdiagnosis. In 2015, the EMHG published a revision of the guidelines for the investigation of MHS. This updated guideline provided a detailed patient referral criteria and clinical interpretation of IVCT results ( https://www.emhg.org/testing-for-mh-1 ).


Only a small number of MHS families have been investigated extensively in North America by phenotyping, linkage analysis, and screening of specific genes. Collaborative protocols over the past several years between MH biopsy centers and molecular biologists have screened 209 unrelated MHS subjects for mutations in the RyR1 gene (see Distribution of RyR1 Mutations).


Larach and coworkers reported a 34.8% morbidity rate in 181 MH cases reported to the NAMHR between January 1987 and December 2006. They also reported that the occurrence was more frequent in young males (75%) (median age of 22.0) and 75% of these patients had undergone at least one general anesthetic with no observed signs of MHS. This underscores the complication of determining the prevalence of MHS in the absence of an inexpensive MH diagnostic test.




Fulminant Malignant Hyperthermia


Fulminant MH is rare. Acute episodes of MH depend on four variables: a genetic (perhaps rarely acquired) predisposition, the absence of inhibiting factors, the presence of an anesthetic or nonanesthetic trigger, and the presence of environmental factors that could potentiate the action of one or more of the other three variables.


Anesthetic Triggering


Anesthetic drugs that trigger MH include ether, halothane, enflurane, isoflurane, desflurane, sevoflurane, and depolarizing muscle relaxants, the only currently used of which is succinylcholine. Desflurane and sevoflurane appear to be less potent triggers than halothane and produce a more gradual onset of MH. The onset may be explosive if succinylcholine is used. MHS swine were traditionally screened by induction with a volatile anesthetic, which led to pronounced hind limb rigidity within 5 minutes, frequently sooner. Prior exercise even an hour before induction of anesthesia increased the severity and hastened the onset of rigidity in swine. Similarly, in the new knock-in mouse models, the onset of limb rigidity after commencing exposure to volatile anesthetics is very rapid. There are also several modifying factors that are more likely to be present in humans than in pigs or mice and can alter (or even prevent) the onset of clinical MH. Mild hypothermia and preadministration of barbiturates, tranquilizers, propofol, or nondepolarizing neuromuscular blockers delay or prevent the onset of MH in MHS humans, thus making them respond less predictably than swine or MH knock-in mice. There have been many instances in which fulminant MH has been reported in patients who have previously tolerated potent triggers without difficulty. The reason behind why this occurs is unknown, but it is likely to be related to prior or concurrent administration of drugs that prevent or delay onset of the syndrome, as described earlier, or unknown environmental influences that help provoke the positive incident. Thus onset of the syndrome in humans is extremely variable both in initial symptoms and in the time of onset of the syndrome. Its onset is so variable that making the diagnosis in the setting of a clinical anesthetic can be quite difficult. Although not perfect, the clinical grading scale developed by Larach and colleagues is a useful way for clinicians to retrospectively determine whether a patient who responded abnormally to anesthesia is in any way likely to actually have had a clinical MH episode. However, MH is most easily diagnosed prospectively by vigilance, recognizing its signs and symptoms, and knowing how to treat the syndrome.


The two classic clinical manifestations of fulminant MH syndrome may start in one of the following two scenarios.



  • 1.

    Rigidity after induction with thiopental and succinylcholine, but successful intubation, followed rapidly by the symptoms listed after scenario 2.


  • 2.

    Normal response to induction of anesthesia and uneventful anesthetic course until onset of the following symptoms:



  • Unexplained sinus tachycardia or ventricular arrhythmias, or both



  • Tachypnea if spontaneous ventilation is present



  • Unexplained decrease in O 2 saturation (because of a decrease in venous O 2 saturation)



  • Increased end-tidal PCO 2 with adequate ventilation (and in most cases unchanged ventilation)



  • Unexpected metabolic and respiratory acidosis



  • Central venous desaturation



  • Increase in body temperature above 38.8°C with no obvious cause



The usually muted onset of MH (scenario 2) is in most cases detected quickly by the development of tachycardia, increased levels of expired CO 2 , and muscle rigidity. It can be delayed for several reasons and may not be overt until the patient is in the recovery room. Once initiated, the course of MH can be rapid. When clinical signs such as increased expired CO 2 , muscle rigidity, tachycardia, and fever suggest MH, more than one abnormal sign must be observed before making the diagnosis because according to a metaanalysis of many reported cases, a single adverse sign does not usually indicate MH. The mechanism by which anesthetics and depolarizing muscle relaxants trigger MH is unsolved, but it cannot be ignored that they are etiologic agents and that early diagnosis is critical for successful treatment.


Nonanesthetic Malignant Hyperthermia


MH can be triggered by stress such as exercise and overheating, known as “awake” MH. Numerous anecdotal reports of MH-like episodes in humans after stressful situations were reported. Measurement of plasma catecholamine levels during exercise showed no differences between MHS and normal individuals. Therefore it is unlikely that these responses were provoked by sympathetic overdrive or catecholamine surge.


Wappler and associates reported RyR1 mutations in three of twelve unrelated patients with exercise-induced rhabdomyolysis (ER); and 10 of those same 12 patients produced abnormal contracture response with IVCT. One had an equivocal response. In susceptible swine, environmental stress such as exercise, heat, anoxia, apprehension, and excitement triggers fulminant MH (see History). These responses are related to muscle movement or to increased temperature. Increased ambient temperature triggers fulminant MH in four strains of heterozygous MH mice and in two homozygous strains. Epidemiologic studies have shown that exercise-induced symptoms, including rhabdomyolysis, may occur more frequently in MHS patients ; and an Arg401Cys RyR1 mutation was present in three cases of exercise-induced rhabdomyolysis. Other reports are largely anecdotal and relate heat stroke, sudden and unexpected death, unusual stress and fatigue, or myalgias to possible “awake” MH episodes. Stresses associated with these episodes include exercise and environmental exposure to volatile nonanesthetic vapors. In the United States, MHAUS provided recommendations of adverse effects of heat and exercise in relation to MHS.




Malignant Hyperthermia–Associated Syndromes


Masseter Spasm (“Thiopental-Succinylcholine or Halothane-Succinylcholine Rigidity”)


A masseter spasm or trismus is defined as jaw muscle rigidity in association with limb muscle flaccidity after the administration of succinylcholine. The masseter and lateral pterygoid muscles contain slow tonic fibers that can respond to depolarizing neuromuscular blockers with a contracture. This is manifested clinically on exposure to succinylcholine as an increase in jaw muscle tone, and was well defined by van der Spek and associates. There is a spectrum of responses, for example, a tight jaw that becomes a rigid jaw and then a very rigid jaw ( Fig. 35.3 ). This jaw rigidity may occur even after pretreatment with a “defasciculating” dose of a nondepolarizing relaxant. If there is rigidity of other muscles in addition to trismus, the association with MH is absolute; anesthesia should be halted as soon as possible and treatment of MH begun.




Fig. 35.3


Succinylcholine usually increases jaw muscle tone slightly. In some patients this increase is moderate, and in very few, the effect is extreme (i.e., “jaws of steel”). As many as 50% of this latter group may be susceptible to malignant hyperthermia (MH) . Somewhere in the area of the declining curve is the boundary for the MH population.


However, in more than 80% of patients with trismus but no rigidity of other muscles, it is a variant found in normal patients. If trismus occurs, proper monitoring should include end-expired CO 2 , examination for pigmenturia, and arterial or venous blood sampling for CK, acid-base status, and electrolyte levels, particularly potassium. Although scientifically unproven, it is thought that the initial tightness of the jaw and its duration may predict the gravity of the response. MHAUS recommends following CK and urine myoglobin for 36 hours with 6-hour intervals. Patients with masseter spasm should be observed closely for at least 12 hours.


Core Myopathies


CCD is a rare hereditary disease. It was first reported in 1956 by Magee and Shy. A recent population study in northern England revealed a prevalence of 1:250,000. In 1971, Engel and colleagues reported a related congenital myopathy, multicore disease. Subsequently, various designations of the terms were reported for the variations of the disease, including minicore myopathy and multiminicore myopathy. MmD is now the most official term for these variations sponsored by the European Neuromuscular Centre.


As mentioned, most CCD cases are due to dominant missense mutations in the RyR1 gene. Clinically, CCD patients present with muscle weakness of variable degree and histologically with central cores in the skeleton muscle type I fibers. MmD is considered a recessively inherited myopathy with severe axial weakness, while respirator, bulbar, and extraocular muscles are commonly affected. MmD has a heterogenous genetic association with a recessive mutation in the SEPN1 gene on chromosome 1p36 and in the RyR1 gene. Both type 1 and type 2 fibers may be affected.


Serum CK levels in CCD patients are often normal but may be elevated up to 6 to 14 times in rare cases. Muscle ultrasound often demonstrates increased echogenicity in the quadriceps muscle with relative sparing of the rectus muscle. This characteristic pattern of selective involvement can also be seen on the muscle MRI and has been reported in the patients with typical CCD, which seems to be distinctive to conditions linked to RyR1 locus.


The relationship between CCD and MHS is complex. A positive IVCT test has been confirmed in many patients with CCD, whereas MHS has been excluded in some. In consideration of the strong link and potential risk, it is advisable to consider all patients with CCD at risk for MH unless the patient has a negative IVCT. Although MHS has not been reported in SEPN1 -related myopathies, it is prudent to apply a nontriggering approach to MmD patients, given the potential risk in RyR1 -related MmD. Clinical MH reactions have been reported in MmD patients.


King-Denborough Syndrome


To address the KDS, we first introduce Noonan syndrome, an autosomal dominant condition involving the face, cardiovascular, hematological, and skeletal systems. Named after Dr. Jacquline Anne Noonan, a pediatric cardiologist, typical Noonan syndrome features delayed puberty, down-slanting or wide-set eyes, hearing loss, low-set or abnormal shaped ears, mild mental retardation (in about 25% of the cases), ptosis, short statue, small penis and undescended testicles in males, pectus excavatum, and a webbed and short neck. The incidence is 1:1000-1:2500 live births. Fifty percent of the patients have protein-tyrosine phosphatase, nonreceptor-type II ( PTPN2 ) mutations. Other genes involved are SOS1, KRAS, RAF1, BRAF, MEK1, and NRAS , and they encode proteins that are part of the Ras (a GTPase)-mitogen activated protein kinase (RAS-MAPK) signaling pathway. Noonan syndrome was recently defined as part of the neuro-cardio-facial-cutaneous syndrome family. An earlier study with a series of 27 patients demonstrated one case of mildly elevated CK despite multiple uneventful cases of general anesthesia with halothane and succinylcholine. Although there is weak evidence for MHS for patients with Noonan syndrome, its resemblances to KDS should raise the concern for the confirmation of the diagnosis. Prevalence of bleeding disorders in Noonan syndrome was reported to be from 20% to 89%, ranging from thrombocytopenia to platelet dysfunction to von Willebrand disease to factor deficiencies. Routine screening including, but not limited to, bleeding history, platelet count, coagulation panel, and factor XI level, was recommended. Hematological consultation becomes appropriate if any of these tests are abnormal. The high palatal arch, dental malocclusion, and the webbed neck of Noonan syndrome make tracheal intubation potentially risky. Nevertheless, odontoid hypoplasia and atlanto-axial instability may result in cervical cord compression. Preoperative cervical spine evaluation is advisable. Right ventricular function needs to be monitored closely because 30% to 50% of the patients with Noonan syndrome have pulmonary stenosis. Regional anesthesia in Noonan patients may be technically challenging due to the prevalence of scoliosis. The spread of local anesthetic can be unpredictable.


KDS features the dysmorphic facial and skeletal abnormalities similar to Noonan syndrome and congenital myopathy with proximal muscle weakness. Sporadic cases have been reported in the literature. The inheritance pattern of the disease is not clear. Elevated baseline CK appears in approximately one half of the KDS patients. A heterozygous A97G point mutation in exon 2 of the RyR1 , causing a substitution of lysine for glutamine at amino acid residue 33 (Lys33Glu), was reported. This substitution creates a major polarity change, from positive to negative, in a known hot spot for an MH causative mutation. Dowling and associates recently identified RyR1 mutation in three out of the four patients with KDS, which supports the hypothesis of its genetic heterogeneity. Given the strong evidence for MHS in KDS patients, MH triggering agents should be avoided for anesthesia on KDS patients.




Diagnosis in the Operating Room and Postanesthesia Care Unit


As stated earlier, fulminant MH is rare, and early signs of clinical MH may be subtle ( Box 35.1 ). These signs must be distinguished from other disorders with similar signs ( Box 35.2 ).



Box 35.1

Clinical Signs of Malignant Hyperthermia


Early Signs





  • Elevated end-tidal CO 2



  • Tachypnea and/or tachycardia



  • Masseter spasm if succinylcholine has been used



  • Generalized muscle rigidity



  • Mixed metabolic and respiratory acidosis



  • Profuse sweating



  • Mottling of skin



  • Cardiac arrhythmias



  • Unstable blood pressure



Late Signs





  • Hyperkalemia



  • Rapid increase of core body temperature



  • Elevated creatine phosphokinase levels



  • Gross myoglobinemia and myoglobinuria



  • Cardiac arrest



  • Disseminated intravascular coagulation




Box 35.2

Conditions and Disorders that May Mimic Malignant Hyperthermia





  • Anaphylactic reaction



  • Alcohol therapy for limb arteriovenous malformation



  • Contrast dye injection



  • Cystinosis



  • Diabetic coma



  • Drug toxicity or abuse



  • Elevated end-tidal CO 2 due to laparoscopic operation



  • Environmental heat gain more than loss



  • Equipment malfunction with increased carbon dioxide



  • Exercise hyperthermia



  • Freeman-Sheldon syndrome



  • Generalized muscle rigidity



  • Heat stroke



  • Hyperthyroidism



  • Hyperkalemia



  • Hypokalemic periodic paralysis



  • Hypoventilation or low fresh gas flow



  • Increased ETCO 2 from laparoscopic surgery



  • Insufficient anesthesia and/or analgesia



  • Malignant neuroleptic syndrome



  • Muscular dystrophies (Duchenne and Becker)



  • Myoglobinuria



  • Myotonias



  • Osteogenesis imperfecta



  • Pheochromocytoma



  • Prader-Willi syndrome



  • Recreational drugs



  • Rhabdomyolysis



  • Sepsis



  • Serotonin syndrome



  • Stroke



  • Thyroid crisis



  • Ventilation problems



  • Wolf-Hirschhorn syndrome




When the diagnosis is obvious (i.e. , fulminant MH or succinylcholine-induced rigidity with rapid metabolic changes), marked hypermetabolism and heat production occur, and there may be little time left for specific therapy to prevent death or irreversible morbidity. If the syndrome begins with slowly increasing end-tidal CO 2 (defined earlier), specific therapy can await a complete clinical workup before treatment. In general, MH is not expected to occur when no triggers are administered (see “Anesthesia for Susceptible Patients”). However, several confirmed fulminant nonanesthetic cases of MH that resulted in death have been reported (see “Nonanesthetic Malignant Hyperthermia”).


When volatile anesthetics or succinylcholine are used, MH should be suspected whenever there is an unexpected increase in end-tidal CO 2 (ETCO 2 ), undue tachycardia, tachypnea, arrhythmias, mottling of the skin, cyanosis, muscle rigidity, sweating, increased body temperature, or unstable blood pressure. If any of these occur, signs of increased metabolism, acidosis, or hyperkalemia must be sought. The most common cause for sudden ETCO 2 during general anesthesia and sedation is hypoventilation. Increased minute ventilation should be able to correct such a problem.


The diagnosis of MH can be supported by the analysis of arterial or venous blood gases which demonstrates a mixed respiratory and metabolic acidosis; however, the respiratory component of acidosis may be predominate in the very early stage of the onset of fulminant MH. O 2 and CO 2 change more markedly in the central venous compartment than in arterial blood; therefore end-expired or venous CO 2 levels more accurately reflect whole-body stores. Venous CO 2 , unless the blood drains an area of increased metabolic activity, should have PCO 2 levels of only about 5 mm Hg greater than that of expected or measured PaCO 2 . In small children, particularly those without oral food or fluid for a prolonged period, the base deficit may be 5 mEq/L because of their smaller energy stores.


Any patient suspected of having an MH episode should be reported to the North American MH Registry via the adverse metabolic/muscular reaction to anesthesia (AMRA) report available from the website at http://anest.ufl.edu/namhr/namhr-report-forms/ .


Treatment


Acute management for MH can be summarized as follows:



  • 1.

    Discontinue all triggering anesthetics, maintain intravenous agents, such as sedatives, opioids, and nondepolarizing muscular blockers as needed, and hyperventilate with 100% oxygen with a fresh flow to at least 10 L/min. With increased aerobic metabolism, normal ventilation must increase. However, CO 2 production is also increased because of neutralization of fixed acid by bicarbonate; hyperventilation removes this additional CO 2 .


  • 2.

    Administer dantrolene rapidly (2.5 mg/kg intravenously [IV] to a total dose of 10 mg/kg IV) every 5 to 10 minutes until the initial symptoms subside.


  • 3.

    Administer bicarbonate (1-4 mEq/kg IV) to correct the metabolic acidosis with frequent monitoring of blood gases and pH.


  • 4.

    Control fever by administering iced fluids, cooling the body surface, cooling body cavities with sterile iced fluids, and if necessary, using a heat exchanger with a pump oxygenator. Cooling should be halted at 38°C to prevent inadvertent hypothermia.


  • 5.

    Monitor and treat arrhythmia. Advanced cardiac life support protocol may be applied.


  • 6.

    Monitor and maintain urinary output to greater than 1 to 2 mL/kg/h and establish diuresis if urine output is inadequate. Administer bicarbonate to alkalinize urine to protect the kidney from myoglobinuria-induced renal failure.


  • 7.

    Further therapy is guided by blood gases, electrolytes, CK, temperature, muscle tone, and urinary output. Hyperkalemia should be treated with bicarbonate, glucose, and insulin, typically 10 units of regular insulin and 50 mL of 50% dextrose for adult patients. The most effective way to lower serum potassium is reversal of MH by effective doses (ED) of dantrolene. In severe cases, calcium chloride or calcium gluconate may be used.


  • 8.

    Recent data demonstrated that magnesium level could be a prerequisite for dantrolene efficacy in managing MH crisis.


  • 9.

    Analyze coagulation studies (e.g. , international normalized ratio [INR], platelet count, prothrombin time, fibrinogen, fibrin split, or degradation products).


  • 10.

    Once the initial reaction is controlled, continued monitoring in the intensive care unit for 24 to 48 hours is usually recommended.



Adequate personnel support is critical to the successful management of such a crisis. Discontinuation of the trigger may be adequate therapy for acute MH if the onset is slow or if exposure was brief. Changing the breathing circuit and CO 2 absorbent can be time-consuming. However, application of activated charcoal filters may rapidly reduce the volatile anesthetic concentration to an acceptable level in less than2 minutes, if they are readily available.


Dantrolene used to be packaged in 20-mg bottles with sodium hydroxide for a pH of 9.5 (otherwise it will not dissolve) and with 3 g of mannitol (converts the hypotonic solution to isotonic). The initial dose should be 2.5 mg/kg dantrolene reconstituted in sterile water and administered intravenously. Dantrolene must be reconstituted in sterile water rather than salt solutions or it will precipitate. It has been shown that prewarming of sterile water may expedite the solubilization of dantrolene compared to water in ambient temperature. In 2009, a newer, rapid soluble lyophilized powder form of dantrolene became available for intravenous use. It reconstitutes in less than a minute which is much faster than the older version. The higher dosing capacity, 250 mg per vial, of the newer version of dantrolene also reduces the storage space with a similar recommended shelf life as the older versions.


In awake, healthy volunteers, the maximum twitch depression occurs at a dantrolene dose of 2.4 mg/kg. Therefore it is not surprising that at therapeutic concentrations, dantrolene may prolong the need for intubation and assisted ventilation. Brandom and associates reviewed the complications associated with the administration of dantrolene from 1987 to 2006 using the dataset in the NAMHR via the AMRA reports and found that the most frequent complications of dantrolene were muscle weakness (21.7%), phlebitis (9%), gastrointestinal upset (4.1%), respiratory failure (3.8%), hyperkalemia (3.3%), and excessive secretions (8.2%). Given its high pH, it is advisable to administer dantrolene through a large bore IV line. It has been demonstrated that dantrolene interferes with EC coupling of murine intestinal smooth muscle cells, rat gastric fundus, and colon, which in part explains its gastrointestinal side effect. Caution should be used when ondansetron is to be used in this setting. As a serotonin antagonist, ondansetron may increase serotonin at the 5-HT 2A receptor in the presynaptic space. In MHS individuals, agonism of 5-HT 2A receptor may produce a deranged response, precipitating MH.


The clinical course will determine further therapy and studies. Dantrolene should probably be repeated at least every 10 to 15 hours, since it has a half-life of at least 10 hours in children and adults. The total dose of dantrolene that can be used is up to 30 mg/kg in some cases. Recrudescence of MH can approach 50%, usually within 6.5 hours. When indicated, calcium and cardiac glycosides may be used safely. They can be lifesaving during persistent hyperkalemia. Slow voltage-gated calcium channel blockers do not increase porcine survival. Instead, a recent study by Migita demonstrated that calcium channel blockers, including dihydropyridine (i.e. , nifedipine), phenylalkylamine (i.e. , verapamil), and benzothiazepine (i.e. , diltiazem), led to increased [Ca 2+ ] i in human skeletal muscle cells. Interestingly, the potency of such calcium release is correlated with the number of binding sites on DHPR (i.e., nifedipine > verapamil > diltiazem). Clinical doses of dantrolene were only able to attenuate 20% of the nifedipine-induced [Ca 2+ ] i surge. Current recommendations of MHAUS discourage the use of calcium channel blockers in the presence of dantrolene because they can worsen the hyperkalemia resulting in cardiac arrest. Although administration of magnesium sulfate could not prevent the development of MH and did not influence the clinical course in succinylcholine-induced MH, recent data suggested that dantrolene might require magnesium to arrest the course of MH triggered by halothane. Permanent neurologic sequelae, such as coma or paralysis, may occur in advanced cases, probably because of inadequate cerebral oxygenation and perfusion for the increased metabolism and because of the fever, acidosis, hypo-osmolality with fluid shifts, and potassium release.


For MH cases diagnosed in the ambulatory surgical centers, guidelines have been recently proposed for the transferring of care to receiving hospital facilities. Although it is preferable that immediate treatment and stabilization of the patient be achieved onsite, several factors need to be considered before implementation of a transfer plan, which include capabilities of the available professionals at the initial treatment and receiving facilities, clinical best interests of patients, and capabilities of the transfer team. The validity of stocking dantrolene in ambulatory surgery centers was confirmed with a cost-effectiveness analysis.




Anesthesia for Susceptible Patients


Safe anesthetics consist of nitrous oxide, barbiturates, etomidate, propofol, opiates, tranquilizers, and nondepolarizing muscle relaxants. Potent volatile anesthetics and succinylcholine must be avoided, even in the presence of dantrolene. There are anecdotal reports that some human patients have experienced a hypermetabolic state despite these precautions, but they have always responded favorably to the administration of intravenous dantrolene. Preoperative dantrolene is never needed because the use of nontriggering agents is almost always associated with uneventful anesthesia. Regional anesthesia is safe and may be preferred. Amide anesthetics such as lidocaine were once considered dangerous in susceptible patients because they were thought to induce or worsen muscle contractures in vitro as a result of their effect of increasing calcium efflux from the SR. Porcine and human studies have consistently demonstrated the lack of danger of amide anesthetics.


Before being used for MHS patients, anesthetic machines may be “cleansed” of potent volatile agents by disconnecting or removing the vaporizers from the anesthesia workstation, renewing the CO 2 absorbent, using a new, disposable breathing circuit, and, if possible, a fresh gas hose. If there is no dedicated machine for MHS patients, flushing the anesthesia workstation to less than5 parts per million (ppm) of the volatile anesthetic agents concentration is generally accepted. It may take 10 to 104 min with different machines. This preparation process also should be directed based on the halogenated volatile agents that have been used. Jones and colleagues demonstrated that desflurane required longer purge time than sevoflurane on both the Datex-Ohmeda Aestiva and Aisys machines. Application of activated charcoal filters have been shown to successfully accelerate the process of cleansing. Such filters should be placed on both the inspiratory and expiratory limbs of the anesthesia machine with replacement of a new set every 60 minutes on patients who are exhaling volatile anesthetics. MHAUS recommends flushing and preparing the anesthesia workstation according to the manufacturer’s recommendations or published studies. During the case, lowering the fresh gas rate after the washout period may allow the concentration of volatile anesthetic agents to reaccumulate. Fresh gas flow should be kept to at least 10 L/min to avoid this rebound.


It is important to be aware that the National Institute for Occupational Safety and Health issued “Criteria for a Recommended Standard- Occupational Exposure to Waste Anesthetic Gases (WAGs) and Vapors.” No worker is exposed to halogenated anesthetic agents at concentrations greater than 2 ppm when used alone or greater than 0.5 ppm when used in combination with nitrous oxide over a sampling period not to exceed 1 hour. Anesthetic gas machines, non-rebreathing systems, and T-tube devices shall have an effective scavenging device that collects all WAGs. Occupational Safety and Health Administration also has guidelines for workplace exposures.


The anesthesiologist should confidently discuss anesthetic care with MHS patients and assure them that all will be done to avoid difficulties with MH and that the appropriate drugs, knowledge, and skills are immediately at hand if any problems occur. Many of these patients have undergone procedures uneventfully, such as dental analgesia and obstetric anesthesia, before the diagnosis of susceptibility was made. The patient can enter the therapeutic environment in a reassured, relaxed, and comfortable state. Outpatient procedures are feasible in most environments; the time of discharge depends on the usual outpatient criteria.


Any facility using MH triggers on an inpatient or outpatient basis should have dantrolene available immediately. The current recommendation by MHAUS to stock 36 vials of 20 mg dantrolene Dantrium/Revonto is based on dantrolene needed to treat an MH crisis on a 70-kg patient. FDA approved the Ryanodex in 2014. Administration of three vials of 250 mg Ryanodex injectable suspension is the alternative preparation plan.


Evaluation of Susceptibility


Evaluation of susceptibility includes a history and physical examination to detect any subclinical abnormality. A genealogy with specific information about anesthetic exposure and agents can estimate the likelihood of exposure to triggering agents. Blood CK values, when determined in a resting, fasting state without recent trauma, reflect muscle membrane stability. When the CK level is elevated in a close relative of a person with known MHS, the relative may be considered to have MHS without contracture testing. If the CK level is normal on several occasions, there is no predictive value, and contracture studies are necessary. The patient must travel to the test center for a surgical biopsy to ensure viability and accurate results. Muscle biopsy contracture studies, performed at about 30 centers around the world, involve exposure of the muscle biopsy sample to halothane, caffeine, and, in the North American test, to halothane plus caffeine. Sensitivity to 4-chloro- m -cresol or ryanodine have also been used by some centers. It is also important to note that contracture responses are sometimes positive in patients with myopathies that bear no direct relationship to MH and therefore may not indicate susceptibility. Dantrolene must be avoided before biopsy because it masks the response to contracture-producing drugs. After a patient is diagnosed as being MHS, DNA testing for mutations should follow. When a mutation is detected, other relatives with that mutation should be considered to be MHS without the need for an invasive contracture test, and they need not travel to a testing center (see Genetics).


MHS patients and all patients who are not biopsy tested, but who present with a clinical picture that suggests a high probability for MHS, should be given advice. Precautions are necessary in regard to general anesthesia, and triggers include all potent volatile agents and succinylcholine. Awake episodes are uncommon, and if not experienced before diagnosis, they are an unlikely problem. The true predictive value (i.e. , percentage of positive results that are true positives) or efficiency (i.e. , percentage of all results that are true, whether positive or negative) of contracture testing in determining susceptibility in the general population cannot be estimated because of the selection process that has been used to date for testing (i.e. , limited to those with anesthesia reactions who do not have any other muscle disease pathology). False-positive results from cautious interpretation or decreased specificity are masked because the patient will never be exposed to triggering agents. A promising innovative in vivo human application involves physiologically based microdialysis infusion of caffeine or halothane into muscle of MHS patients to trigger exaggerated localized changes in acid-base balance. White blood cells express RyR1-MHS and provide a substrate for a less invasive analysis for susceptibility but have the limitation that not all causative mutations are expressed in the white blood cells. Nuclear magnetic resonance has promise, but to date it has been difficult to standardize a stress, such as forearm ischemia, that can differentiate susceptibles from normals.


For anesthesia assessment of the non-MHS pregnant patient carrying a potential MHS fetus, the parturient should be treated as MHS until the fetus is delivered. For emergencies in such patients, the use of succinylcholine, although little of the drug crosses the placenta, is controversial.




Multiple Sclerosis


Multiple sclerosis (MS) is an autoimmune disorder characterized by T-cell–mediated autoantibodies against myelin and a subsequent inflammatory response within the central nervous system (CNS: brain and spinal cord), where it primarily affects the optic nerve, the corticospinal tract, or posterior columns. Thus MS is a disorder of the myelinated part of the axon that leads to secondary nerve conduction failure. It is characterized by sensitization of the peripheral leukocytes to the myelin antigen and a subsequent inflammatory response, monocytic and lymphocytic perivascular cuffing, and glial scars with plaque formation within the central nervous system, especially in the periventricular white matter. The disease affects mainly women, primarily between 20 and 40 or 45 and 60 years of age. Although the etiology is unknown, it has been speculated that MS is caused by environmental factors combined with a genetic predisposition. Naturally, researchers have focused on identifying key events and the genetic origin of the disorder to provide diagnostic and possibly also therapeutic tools for the management of MS patients.


Its clinical course is characterized by exacerbations and remissions in most patients, but continuous neurological deterioration has been reported in up to 10% of cases (primary progressive MS). Patients with MS frequently report paresthesias, muscle weakness, and sensory disturbances. Acute symptoms, which are related to the site and extent of sclerosing CNS plaques, commonly include visual disturbances (diplopia, blurring, and field cuts), sensory abnormalities with numbness and paresthesia, pain, and electric shock sensation that radiates down the spine and into the limbs upon flexion of the neck (Lhermitte sign). Cranial nerve dysfunction, ataxia, and bladder and bowel disturbances are also common. Typically, there is a localized or, late in the course of disease, generalized muscle weakness with the legs affected more than the arms. Chronic symptoms can also include spastic paraparesis, appendicular tremor, psychiatric disturbances such as depression or euphoria (la belle indifference), and dementia. In severe cases, respiration may be involved with the development of hypoxemia. As a rule, symptoms are closely related to the site affected within the CNS, and the number of symptoms is related to the extent of sclerosing CNS plaques. Notably, MS can be associated with impaired autonomic function, and hence an increased risk for exaggerated hemodynamic response to anesthetic induction agents, vasodilators, and sympathomimetic drugs.


Diagnosing MS after a single, acute remitting clinically isolated syndrome is discouraged, whereas repeated attacks with increased CSF IgG and multifocal MRI abnormalities are strongly supportive of the diagnosis. Acute attacks are treated with various combinations of immunosuppression modalities including glucocorticoids or plasma exchange therapy, which have been shown to increase the rate of recovery but not the overall level of recovered function. Disease progression can, nevertheless, be modified with a novel humanized CD20 monoclonal antibody (ocrelizumab) in patients with primary-progressive or relapsing-remitting forms of MS. Other immunomodulatory treatment options include interferon β1a or glatiramer acetate (a mixture of polypeptides synthesized to mimic myelin basic protein) in individuals with the relapsing-remitting MS, fingolimod, teriflunomide, or natalizumab, and the antineoplastic agent mitoxantrone. Teriflunomide is associated with hepatic injury, while mitoxantrone can be associated with cardiomyopathy. These patients may also receive treatments aimed at reducing spasticity (baclofen and benzodiazepines), as well as anticonvulsants or propranolol for tremor, oxybutynin and propantheline for bladder spasticity, and SSRIs or other antidepressive agents for mood disorders.


Anesthetic Considerations


It has been speculated that general anesthesia and surgery may increase the risk for aggravation of MS. Presently, there is no general consensus on this matter, and patients should therefore be informed of the potential for aggravated symptoms in the postoperative period. In general, preoperative chronic immunosuppressive medication should be continued during the perioperative period. Patients with MS are sensitive to physical (pain, fever, infection) and emotional stress, which makes it more likely that symptoms will be intensified in the perioperative period. Increased body temperature is often cited as an offending mechanism, possibly by causing a complete block of conduction in demyelinated nerves. Body temperature should therefore be closely monitored and controlled during the perioperative period. Great care must be exercised to minimize changes in fluid homeostasis, and central hemodynamics (preload, afterload) and to maintain respiration. Although intravenous induction agents and volatile anesthetics have been used safely, it is wise to avoid administering depolarizing neuromuscular blocking drugs to MS patients. MS-induced denervation, or misuse myopathy, may lead to a risk for succinylcholine-induced hyperkalemia, which can result in fatal cardiac arrhythmias. Nondepolarizing neuromuscular blockers are safe to use but should be dosed cautiously as both prolonged responses (increased sensitivity in patients with preexisting muscle weakness) and resistance to these NMBAs have been reported. The use of rocuronium with sugammadex to ensure full reversal has been suggested as a safe alternative. It is speculated that the demyelination associated with MS renders the spinal cord susceptible to the neurotoxic effects of the local anesthetics. Epidural application of low concentrations of local anesthetics has, nevertheless, been successfully used in MS patients. Spinal anesthesia, on the other hand, has been implicated in postoperative exacerbations of symptoms in MS. As the blood–brain barrier may also be damaged by demyelination, spinal anesthesia is usually not recommended for these patients. A recent metaanalysis of 37 reports found that the MS symptoms were worsened in 10 of 231 patients, but despite the association no clear cause-effect relationship could be identified. Notably, postpartum worsening of MS symptoms is noted in 20% of females. The need for postoperative care is dependent on the preoperative symptoms, type of surgery, and status of the patient at the end of the surgical procedure. In this context, MS patients with severe weakness and respiratory distress, including pharyngeal dysfunction, may need extended postoperative care, such as noninvasive respiratory support and intense physiotherapy, to avoid further impairment of their pulmonary function ( Box 35.3 ).



Box 35.3

Perioperative Considerations for Patients with Multiple Sclerosis




  • 1.

    Thoroughly inform the patient and family of the natural course of MS, and the risk for perioperative worsening of symptoms


  • 2.

    Continue preoperative immunosuppressive therapy


  • 3.

    Type of general anesthetics is unlikely to affect the course of disease


  • 4.

    Minimize perioperative changes in fluid homeostasis and hemodynamics


  • 5.

    Monitor body temperature closely, avoid hyperthermia


  • 6.

    It is reasonable to avoid depolarizing neuromuscular blocking agents (NMBAs)


  • 7.

    Nondepolarizing NMBAs can be used, but should be dosed cautiously with monitoring of the neuromuscular transmission


  • 8.

    Epidural anesthesia has been used successfully, but spinal anesthesia is usually not recommended


  • 9.

    Consider extended postoperative care in a monitored setting if patient has severe preoperative weakness or respiratory compromise






Motor Neuron Disorders


Motor neuron disorders involve either the upper or the lower motor neurons of the cerebral cortex, brainstem, and spinal cord. Some forms are mixed, whereas others have predominately upper or lower motor neuron involvement. Amyotrophic lateral sclerosis (ALS, also known as Lou Gehrig’s disease) is the most common disease within this group and involves both upper and lower motor neurons. Other examples of motor neuron disease are Kennedy disease (spinobulbar muscular atrophy), Friedreich ataxia (mixed upper and lower motor neurons), and spinal muscular atrophy (lower motor neurons).


ALS is characterized by degeneration of the anterior horn α-motoneurons in the spinal cord and brainstem motor nuclei, as well as the primary descending upper motor neurons of the corticospinal tract. Degenerative loss of these neurons leads to progressive muscle weakness, muscle atrophy, and loss of neuronal mass in these locations. Patients present with gradually spreading focal weakness and muscle atrophy (typically of the hands), spasticity, and hyperreflexia of lower extremities. Dysarthria and dysphagia, tongue atrophy, and fasciculations may also occur. Progressive weakness can lead to respiratory failure and death. Sensory functions, including intellectual capacity and cognition, as well as bowel and bladder function, are not usually affected in ALS.


ALS has an incidence of about 2 in 100,000, and onset of the disease usually takes place around 40 to 50 years of age, with males more often affected than females. Most cases are sporadic, but rare familial forms (autosomal dominant and recessive forms) do exist. The underlying mechanism or mechanisms for this selective and progressive motor neuronal death are thus far unclear, but it has recently been suggested that superoxide dismutase (SOD) mutations may have a key role in the increased formation of free radicals seen in subsets of patients. SOD is an important antioxidant and its mutation can lead to decreased clearance of free radicals, increased oxidative stress, and mitochondrial dysfunction. Most familial forms are associated with the mutation of C9ORF72 on 9p21, TDP43, FUS, and VCP genes. The diagnosis is made by electrophysiology (electromyography [EMG] and electroneurography), neurologic examination, MRI imaging, and CSF analysis, which demonstrates early spastic weakness of the upper and lower extremities, typical subcutaneous muscle fasciculations, and bulbar involvement affecting pharyngeal function, speech, and the facial muscles. No curative treatment is currently available, and patients are therefore treated symptomatically. Riluzole, a glutamate release inhibitor, may provide neuroprotection and extend survival in these patients. More recently, the antioxidant edaravone was shown to reduce the decline in daily functioning associated with ALS. Patients may also receive spasmolytic and analgesic agents. Those with advanced disease will ultimately require tracheostomy and gastrostomy surgeries and other supportive treatments including mechanical ventilation.


Anesthetic Considerations


Bulbar involvement in combination with respiratory muscle weakness leads to a risk for aspiration and pulmonary complications. Notably, these patients may have increased sensitivity to the respiratory depressant effects of sedatives and hypnotics. There are reports of sympathetic hyperreactivity and autonomic failure. Sympathetic hyperreactivity and autonomic dysfunction, often manifested as orthostatic hypotension and resting tachycardia but also significant hypotension or even pulseless electrical activity upon anesthesia induction, have been reported, and should be considered during the perioperative management of these patients. Succinylcholine should be avoided because of the risk for hyperkalemia as a result of denervation and immobilization. Nondepolarizing NMBA may cause prolonged and pronounced neuromuscular blockade and hence should be used with great caution. General anesthesia may be associated with exaggerated ventilatory depression. Regional anesthesia is also often avoided for fear of exacerbating disease symptoms. Both general and epidural anesthesia have, however, been successfully administered to these patients without reported complications ( Box 35.4 ).



Box 35.4

Perioperative Considerations for Patients with Amyotrophic Lateral Sclerosis




  • 1.

    Exaggerated respiratory depression and sensitivity to sedatives and hypnotics


  • 2.

    Higher risk for aspiration and pulmonary complications


  • 3.

    Autonomic dysfunction with risk for hemodynamic instability


  • 4.

    Avoid depolarizing neuromuscular blocking agents (NMBAs) (risk for hyperkalemia); nondepolarizing NMBAs may cause prolonged and profound neuromuscular blockade


  • 5.

    General and epidural anesthesia have been successfully administered; spinal anesthesia is often avoided






Guillain-Barré Syndrome


Guillain-Barré syndrome or acute inflammatory demyelinating polyradiculopathy is an acute inflammatory polyneuritis that is triggered by humoral and cell-mediated autoimmune response to a sensitizing event. Although the etiology is unknown, in many cases a timely association with a viral (influenza-like) or bacterial infection or even lymphomatous disease can be demonstrated. It typically presents as an ascending paralysis characterized by symmetric weakness that can vary from mild difficulty with walking to nearly complete paralysis of all extremities, facial, respiratory, and bulbar muscles. Mild variants can present with ataxia, ophthalmoplegia, or hyporeflexia without significant appendicular weakness. Fulminant cases can present with severe ascending weakness leading to complete tetraplegia, and paralysis of cranial nerves and phrenic and intercostal nerves with facial and respiratory muscle weakness necessitating tracheostomy and ventilatory support. Importantly, patients may also have autonomic involvement that could lead to hemodynamic instability and arrhythmias with risk for sudden circulatory collapse and fatal cardiac.


The diagnosis is made after careful neurologic examination such as areflexia and progressive motor weakness, clinical and electrophysiological studies, and CSF analysis. CSF analysis may show a typical increase in CSF protein in combination with a normal cell count, which is a classic sign of the disease. Electromyogram (EMG) and nerve conduction studies may be normal in the early acute period, but characteristic segmental demyelination and reduction of conduction velocity and dispersion or absence of F-waves are usually seen within 1 to 2 weeks.


Management is primarily supportive and includes nutritional support, respiratory support, and measures to prevent aspiration. Early plasma exchange, typically five exchanges with 5% albumin repletion, may mitigate the course but is contraindicated in setting of hemodynamic instability, marked dysautonomia, and active bleeding. Intravenous immunoglobulin (IVIG) is typically administered in the setting of dysautonomia, or if plasmapheresis and exchange transfusion are contraindicated.


Anesthetic Considerations


Cranial nerve paralysis and autonomic dysfunction predispose these patients to an increased risk for aspiration. Aspiration precautions, including decompression of the stomach, should therefore be considered before the induction of anesthesia. Absence of compensatory cardiovascular responses may be associated with exaggerated hypotension at anesthesia induction or in response to hypovolemia. Conversely, laryngoscopy or noxious stimuli can be associated with an exaggerated increase in blood pressure. The hemodynamic instability is typically short-lived and self-limited, but small doses of short-acting and titratable vasoactive medications may be required. Careful hemodynamic monitoring is essential and continuous monitoring of the blood pressure with an arterial catheter is often considered. These patients may also exhibit abnormal responses to NMBA; succinylcholine should not be used because of the risk of hyperkalemia. Nondepolarizing muscle relaxants are not contraindicated but should be avoided as a result of the increased sensitivity and risk for prolonged muscle weakness in the postoperative period. The risk for autonomic dysfunction, respiratory failure, and aspiration may require assisted or mechanical ventilation, even in the postoperative period. If these agents are used, the neuromuscular transmission should be closely monitored with a nerve stimulator as both resistance and sensitivity to these agents have been reported. Great care should be taken to maintain circulatory stability, including adequate cardiac preload and afterload. Careful hemodynamic monitoring is therefore essential in these patients.


Regional anesthesia is employed by some practitioners but its use remains controversial as it has been reported to cause worsening of neurological symptoms. General anesthesia can be used; however, the combination of general anesthesia and epidural anesthesia is more controversial ( Box 35.5 ).



Box 35.5

Perioperative Considerations for Patients with Acute Inflammatory Demyelinating Polyradiculopathy




  • 1.

    Autonomic dysfunction may be associated with hemodynamic instability and an exaggerated response to anesthesia induction agents, or to stimulating interventions such as laryngoscopy


  • 2.

    Depolarizing neuromuscular blocking agents (NMBAs) should be avoided due to an upregulation of the acetylcholine receptors and risk for hyperkalemic response


  • 3.

    Nondepolarizing NMBAs can be used but are commonly also avoided because of the risk for prolonged weakness


  • 4.

    The use of regional anesthesia is controversial and may be associated with worsening symptoms




Critical Illness Polyneuropathy and Critical Illness Myopathy


Despite earlier reports of a rapid development of weakness, muscle atrophy, and polyneuropathy in critically ill patients, it was not until the 1987 report by Bolton and associates that the characteristic widespread axonal degeneration of motor and sensory fibers and the extensive denervation atrophy of limb and respiratory muscle associated with this polyneuropathy were clearly identified. Although the true incidence of critical illness polyneuropathy (CIP) is difficult to determine, critical illness neuropathy and myopathy are believed to affect up to 50% of all patients remaining in the intensive care unit for more than 2 weeks. It is typically manifested as profound symmetric limb weakness, with reduced or absent tendon reflexes and diaphragmatic and intercostal weakness. It affects the lower extremities to a greater extent than upper extremities, and distal muscle groups more severely than the proximal. The autonomic function is not affected and the extraocular eye movements remain intact. In CIP, there is no evidence of neuromuscular junction disorder and the electromyography and nerve conduction study findings are consistent with axonal motor and sensory polyneuropathy, with amplitude reduction of motor and sensory action potentials, and slowed conduction velocities. Serum CK levels are usually normal. Conversely, a sensory nerve action potential is often normal in critical illness myopathy but compound muscle action potentials are diminished and electromyography is consistent with myopathy. Serum CK levels may be elevated. No specific treatments are currently available, and management is supportive with aggressive and early rehabilitation. Use of sedation, paralytics, and corticosteroids should be limited, and aggressive control of hyperglycemia has been suggested to reduce the incidence of CIP by 44%.


Anesthetic Considerations


Anesthetic considerations in CIP patients are similar to those with other acquired neuropathies (see above), and include protection of nerve compression sites, particularly the ulnar and peroneal nerves. Prolonged immobility in critically ill patients is associated with a relative increase in immature acetylcholine receptors that can lead to an insensitivity to nondepolarizing NMBA. Conversely, the sensitivity to depolarizing neuromuscular blockers is increased, with a risk for increased potassium efflux after succinylcholine administration ( Box 35.6 ).



Box 35.6

Perioperative Considerations for Patients with Critical Illness Polyneuropathy




  • 1.

    Particular attention should be made to protect peripheral nerves, in particular the ulnar and peroneal nerves, during positioning of these patients


  • 2.

    Monitor and correct electrolyte and glucose abnormalities


  • 3.

    Steroids have been implicated in the pathophysiology of the disease and should therefore be avoided


  • 4.

    Neuromuscular blocking agents are best avoided altogether, but if needed only nondepolarizing agents should be considered






Hereditary Motor-Sensory Neuropathies, including Charcot-Marie-Tooth Disease


Hereditary motor-sensory neuropathies include a spectrum of peripheral neurologic disorders, among which Charcot-Marie-Tooth (CMT) disease is often listed. They are caused by a specific mutation in one of several myelin genes that result in defects in myelin structure, maintenance, and formation. Hereditary motor-sensory neuropathies have been classified into seven types and multiple subtypes according to the age at onset, mode of inheritance, predominately involved muscle groups, and genotypes. CMT types 1 and 2 are the most common hereditary peripheral neuropathies, with an estimated prevalence of 40 per 100,000. Patients with CMT disease typically experience slow and progressive distal muscle weakness and wasting. Damage to sensory axons may also lead to sensory loss resulting in frequent tripping and falls. Neuropathic pain may develop in some patients. CMT patients usually have normal life expectancy. CMT type 3, also known as Dejerine-Sottas disease, is a very severe condition with an early onset of hypotonia during infancy. Nerve conduction velocity is typically significantly reduced to less than 10 ms. The genetic inheritance pattern for CMT disease is heterogeneous.


Anesthetic Considerations


The anesthesia experience in patients with CMT disease is limited because of the small number of cases. Major considerations include the use of hypnotic agents, muscle relaxants, volatile agents, and neuraxial techniques. CMT type 1 patients have been reported to have significantly increased sensitivity to thiopental at induction that correlates with the severity of both motor and sensory defects. However, total intravenous anesthesia (TIVA) has been performed successfully in a number of cases without any reported problems.


Because of the reduced number of acetylcholine receptors, sensitivity to nondepolarizing muscle relaxants is elevated and the response to succinylcholine is also reduced. Although succinylcholine has been used without adverse effect, the risk of an exaggerated hyperkalemic response may be sufficient to preclude it from being used in patients with suspected muscular denervation. Prolonged neuromuscular blockade with vecuronium has been reported. As a result of the large variety of disabilities in this patient group, careful baseline assessment of neuromuscular status should be conducted before the use of nondepolarizing neuromuscular relaxants. Normal response to atracurium and mivacurium has been demonstrated. Both TIVA and volatile anesthetics have been used safely in CMT patients in a series of cases. Neuraxial techniques for obstetric procedures have been reported to generally be successful in CMT patients. However, the use of regional anesthesia can be controversial given that the possible complications may exacerbate the neurologic symptoms. Similar medicolegal concerns may apply to the surgical and anesthesia positioning of CMT patients because of the sensory deficits and limb deformities.




Duchenne Muscular Dystrophy and Becker Muscular Dystrophy


Duchenne muscular dystrophy (DMD) is the most common and severe type of muscular dystrophy, with an incidence of 1 per 3500 live male births and a total male prevalence of about 50 to 60 × 10 –6 . Becker muscular dystrophy (BMD) is relatively rare and has an incidence of about 1 in 18,000 live male births and a prevalence of 23.8 × 10 –6 . Both DMD and BMD are X-linked recessive diseases. The defect is located on the short arm of the X chromosome at the Xp21 region, which contains the gene for the large protein Dp427, also known as dystrophin. The dystrophin gene is 2500 kilobases long with more than 70 exons. Dystrophin is distributed not only in skeletal, cardiac, and smooth muscle but also in the brain. Because of the large size of the dystrophin gene, spontaneous new mutations are common and account for a third of new cases.


The most common form of mutation is a deletion within the gene (65%-70% of cases of DMD and >80% of BMD). Duplication and point mutations are responsible for the rest. It also appears that there are “hot spots” within the first 20 exons and in the central region of the gene (exons 45-55) where deletion and duplication are likely to occur. Female cases of DMD have been reported with the 45,X and 46,XX karyotypes. The disease mechanism for the female 46,XX karyotype was thought to be preferential loss of the paternal X chromosome by postzygotic nondisjunction and manifestation of the DMD gene from the maternal X chromosome in muscle cells. BMD is usually milder in severity than DMD because disruption of the translation process occurs in the relatively distal part of the gene, which leads to a reduced amount of truncated dystrophin protein.


Dystrophin, along with dystrophin-associated glycoproteins (DAGs), is involved in sarcolemmal stability. Dystrophin is responsible for maintenance of muscle membrane integrity despite the fact that it accounts for only approximately 0.002% of the protein in striated muscle. Dystrophin aggregates and links to actin (at its N terminus) and the DAG complex (at its C terminus) to form a stable structure that interacts with laminin in the extracellular matrix ( Fig. 35.4 ). Lack or dysfunction of dystrophin leads to cellular and membrane instability, with progressive leakage of intracellular components and elevation of creatine phosphokinase (CPK) levels. Eventually, damaged muscle cell units are invaded by macrophages and destroyed. Current study suggests that cytotoxic T cells are probably the culprit. Consequently, clinical pseudohypertrophy of the muscle occurs when the dead muscle shells are replaced by fibrofatty infiltrates. Loss of muscle units accounts for the weakness and contracture.




Fig. 35.4


Diagram of the cell–surface and cytoskeleton protein complex.


Both DMD and BMD are characterized by progressive weakness and wasting of predominantly the proximal musculature. Pseudohypertrophy of the calves and other muscle groups is common. As the more severe of the two diseases, DMD tends to be symptomatic early in life. Seventy-four percent of children with DMD were found to manifest the disease by 4 years of age. DMD patients do not usually begin to walk until they are about 18 months of age or later.


The initial clinical findings include a waddling gait, frequent falling, and difficulty climbing stairs because of proximal muscle weakness in the pelvic girdle. The classic Gower maneuver describes rising from a sitting to a standing position with the help of both arms. Patients may also show weakness in the shoulder girdle and trunk erectors that leads to thoracolumbar scoliosis. The earlier the onset of disease, the more rapid the disease will take its course. In most cases, children with DMD are unable to walk by the age of 9 to 11. Proximal deep tendon reflexes in the upper extremities and patella may also disappear despite the lack of denervation. Nevertheless, the Achilles tendon reflex remains intact even in later stages of the disease. Sixty percent of patients will have pseudohypertrophy of the calves, and thirty percent will have macroglossia. Some may also suffer from pain in the calves with activity.


The intellectual impairment that can be associated with the disease was thought to be related to limitation of educational opportunities. However, with equalization of educational opportunities, psychometric studies have revealed a significantly lower average intelligence quotient in DMD patients than in healthy groups. This implies a possible effect of dysfunctional dystrophin in the brain on learning.


Death in patients with DMD is commonly due to cardiopulmonary compromise in their 30s. BMD is a mild form of DMD. The mutation that causes BMD produces dystrophin that retains partial function. The onset of symptoms occurs in the second or third decade of life. As a result, the life span of BMD patients can reach the early 40s. Pneumonia is the most common cause of death ( Fig. 35.5 ).




Fig. 35.5


Distribution of predominant muscle weakness in different types of dystrophy: (A) Duchenne type and Becker type; (B) Emery-Dreifuss; (C) limb girdle; (D) facioscapulohumeral; (E) distal; and (F) oculopharyngeal.

Redrawn from Emery AE. The muscular dystrophies. BMJ 1998;317:991–995.


The heart is also affected to various degrees, depending on the stage of the disease and the type of mutation. Cardiac degeneration is due to replacement of myocardium by connective tissue or fat, which leads to dilated cardiomyopathy. Cardiac involvement starts early in the course of the disease, although clinical signs are not usually obvious in the early stage. No correlation has been established between the severity of cardiac disease and the severity of skeletal disease. Studies of necropsy have shown that the cardiomyopathy in DMD involves the posterobasal and contiguous lateral left ventricular walls as initial and primary sites of myocardial dystrophy in the absence of small vessel coronary artery disease in these areas. Typical initial manifestations on the electrocardiogram (ECG) in DMD and BMD are sinus tachycardia, tall R waves in the right precordial leads, prominent left precordial Q waves, increased QT dispersion, and inverted T waves from scarring of the posterobasal portion of the left ventricle. Initially, the echocardiography is normal or shows regional wall motion abnormalities in areas of fibrosis. With the spreading of fibrosis, left ventricular dysfunction can be seen and ventricular arrhythmias frequently occur as well. In the final stages of the disease, systolic dysfunction may lead to heart failure and sudden death. Subclinical or clinical cardiac involvement is present in about 90% of DMD/BMD patients, but it is the cause of death in only 20% of DMD and 50% of BMD patients. Angiotensin-converting enzyme inhibitors are recommended in early stages of the disease, and β-blockers may be an additional option if indicated.


Pulmonary insufficiency is a leading cause of morbidity and mortality in DMD. Usually, expiratory muscle function is affected first, because of the early onset of abdominal muscle weakness. By contrast, inspiratory muscle function is relatively preserved in the first decade, implying sparing of the diaphragm. Vital capacity (VC) increases in the first decade because of overall body growth, plateaus in early adolescence, and then declines dramatically as the diaphragmatic weakness progresses. Other measured lung volumes such as inspiratory reserve volume and total lung capacity (TLC) follow the same pattern. A disproportionate loss of VC and TLC relative to the respiratory muscle dysfunction results in part from additional factors, such as altered chest wall and lung mechanics, modifications in the distribution of surfactant, micro-atelectasis, and local fibrosis secondary to recurrent pneumonia. Scoliosis further impairs pulmonary function. On average, for each 10 degrees of thoracic scoliosis curvature, forced vital capacity (FVC) decreases by 4%. In 90% of patients, a curvature of greater than 20 degrees develops 3 to 4 years after they are wheelchair bound. Respiratory failure inevitably occurs in the second decade of life and is the most common cause of death.


Diagnosis and Differential Diagnosis


Chronic elevation of the serum CPK level is a general indication of muscle disease. Three serum tests showing elevated CPK levels obtained one month apart is diagnostic of muscular dystrophy. CPK represents leakage of enzyme from muscle cells and does not correlate with severity of the disease. CPK could reach 50 to 300 times the normal value in early stages of the disease. The level tends to decrease with the loss of muscle mass. Elevation of the MB fraction of CK precludes its use as a marker for cardiac injury. EMG can be supportive of the diagnosis; however, it can be very difficult to perform on children. Muscle biopsy, followed by immunostaining or Western blot analysis for dystrophin, is recommended for diagnostic testing. Multiple polymerase chain reactions are also useful in detecting more than 98% of the existing deletions. The result is usually available within 24 hours, which may render muscle biopsy, the old “gold standard,” obsolete.


Anesthetic Considerations


Patients with DMD and BMD may require anesthesia for muscle biopsy, correction of scoliosis, release of contractures, and exploratory laparotomy for ileus, as well as for dental and obstetric procedures. As the natural course of the disease progresses, the risk of surgery increases concomitant with the increased comorbid conditions associated with the later phase of the disease. However, perioperative complications are not proportional to the severity of the disease. They occur even in mildly affected patients. Consequently, patients should undergo careful preoperative consultation and evaluation.


Fifty to seventy percent of patients with muscular dystrophy demonstrate some cardiac abnormality, although it is clinically significant in only ten percent. Preoperative cardiology assessment with an ECG and echocardiography is essential. Continuous cardiac Holter monitoring is necessary if an arrhythmia is captured on the ECG or if the patient describes symptoms that can be related to cardiac arrhythmias. An echocardiography will demonstrate mitral valve prolapse in 10% to 25% of patients. It may also show posterobasilar hypokinesis in a thin-walled ventricle and a slow relaxation phase with normal contraction characterizing the cardiomyopathy seen in DMD. However, echocardiography may not always reflect the ability of the diseased myocardium to respond to perioperative stress. A stress echocardiography using angiotensin to detect latent heart failure and identify inducible contraction abnormalities has been advocated.


An estimated 30% of deaths in individuals with DMD are due to respiratory causes. Therefore careful preoperative pulmonary assessment is important. Webster demonstrated that the manual muscle strength test has a strong statistical correlation with all of the timed functional tests. Peak expiratory flow was not only easy to perform, but also correlated statistically with all timed functional tests. The correlation was not significant for VC or forced expiratory volume in 1 second (FEV 1 ).


Intraoperatively, in terms of airway management, patients with DMD and BMD may have decreased laryngeal reflexes and prolonged gastric emptying time, which increases the risk for aspiration. Decreased ability to cough up the accumulation of oral secretions predisposes muscular dystrophy patients to postoperative respiratory tract infections. Masseter spasm is also a possible complication during induction of anesthesia in these patients. Preparedness for a difficult airway is necessary, especially in patients with potential airway problems.


Postoperatively, DMD patients are at an increased risk for respiratory compromise. Retrospective reviews have indicated that the incidence of prolonged postoperative ventilation (>36 hours) was greatest in DMD patients who had a preoperative FVC of less than 40% of the predicted value. Preoperative pulmonary function studies are valuable in determining the postoperative course. Patients with a VC of greater than 30% of the predicted value can usually be extubated immediately after surgery. Sleep apnea may also compound the condition and contribute to the development of pulmonary hypertension. Continuous positive airway pressure and bilevel positive airway pressure have been demonstrated to be effective in the management of postoperative respiratory depression. Delayed pulmonary insufficiency may occur up to 36 hours postoperatively despite the apparent recovery of skeletal muscle strength.


Reports have suggested a relationship between DMD/BMD and MH, but this association is not based on good rational grounds. Whereas the risk for an MH mutation in DMD/BMD patients is similar to that of the general population, the incidence of MH-like anesthetic events has been reported to be 0.002 with DMD and 0.00036 with BMD. Unexplained cardiac arrest and acute heart failure have been reported in DMD/BMD patients. Succinylcholine is contraindicated in these patients because of the potential for rhabdomyolysis and hyperkalemia as a result of their unstable sarcolemmal membranes. Succinylcholine-induced hyperkalemia during acute rhabdomyolysis is more likely to result in cardiac arrest and unsuccessful resuscitation than is the potassium efflux resulting from upregulation of acetylcholine receptors in burn patients. Although the use of nondepolarizing muscle relaxants is usually accompanied by an increase in both maximal effect and duration of action, proper and rapid reversal with sugammadex may put some of these concerns to rest. Current off-label experience in infants and children has been favorable. Narcotics can be used, but small incremental dosing and short-acting medications are recommended given the respiratory depression associated with these medications, as well as the reports of inadvertent reactions to volatile anesthetics,


Recently, TIVA has become more popular. However, consideration needs to be given to the myocardial status of the patient when propofol or barbiturates are used because they may lead to profound hypotension and reduced organ perfusion. Regional anesthesia may be a good alternative to general anesthesia because it avoids the risk of triggering agents and respiratory depression and enables the use of local anesthetics for postoperative analgesia. It may also facilitate chest physiotherapy.


Recent breakthroughs in gene therapy are shining new lights into the management of these relatively common disorders. We have not seen reports on anesthesia management for DMD/BMD patients who had received gene therapy.




Limb-Girdle Muscular Dystrophy


Limb-girdle muscular dystrophy (LGMD) is a group of disorders with heterogeneous causes. To date, at least 18 genes have been identified as being responsible for this disease, with 7 being autosomal dominant and 11 autosomal recessive. Mutations within the same gene may result in different phenotypes that sometimes are not consistent with LGMD. Proximal muscle (shoulder or pelvic) girdle weakness is the characteristic feature of this group of diseases. Given the marked genetic heterogeneity, clinical manifestations of the disease vary. Autosomal recessive forms are about 10 times more common than autosomal dominant forms. Fukutin-related protein ( FKRP ) and calpain 3 ( CAPN3 ) gene mutations have been associated with LGMD. In addition, a number of other disorders, not strictly included under LGMD in this classification, may have LGMD-like phenotypes. Sporadic cases of LGMD have been reported in the anesthesia literature. General approaches to these patients are the same as those for DMD/BMD.




Myotonic Dystrophy


Myotonic dystrophy (MD) is an inherited muscular disorder characterized by progressive muscle weakness and wasting. Two types of MD result from a mutation in either the dystrophia myotonica–protein kinase ( DMPK ) gene, located on chromosome 19q13.3 (MD1, also known as Steinert disease), or the CysCysHisCys (CCHC)-type zinc finger, nucleic acid binding protein ( CNBP ) gene, located on chromosome 3q21 (MD2).


The incidence of MD is 1 in 8000. MD1 is by far the most common of the two types and accounts for about 98% of all cases. MD1 is caused by expansion of a CTG trinucleotide repeat in the DMPK gene and is inherited in an autosomal dominant manner. Typical signs and symptoms include muscle weakness and wasting (most prominent in the cranial and distal limb musculature), periodic myotonia, progressive myopathy, insulin resistance, defects in cardiac conduction, neuropsychiatric impairment, cataracts, testicular atrophy, and frontal balding in males. The typical cranial muscle weakness and wasting are manifested not only in the facial, temporalis, masseter, and sternocleidomastoid muscles but also in the vocal cord apparatus. Mitral valve prolapse is found in 20% of patients. The severity of the disease is related to the number of extra trinucleotide repeats. MD1 patients may also have mildly elevated CK levels. Myotonic discharges can be identified on EMG, as well as an inability to relax from a handgrip. During pregnancy, the symptoms may be exacerbated. Uterine atony and a retained placenta may also complicate vaginal delivery. First-degree atrioventricular heart block is a common finding on the ECG before the onset of symptoms.


MD2 is also called proximal myotonic myopathy. Intron 1 of the CNBP gene contains a complex repeat motif, (TG)n(TCTG)n(CCTG)n, and expansion of the CCTG repeat was determined as the cause of MD2. Patients with MD2 suffer from myotonia (90% of those affected), muscle dysfunction (82% weakness, pain, and stiffness), and less commonly, cardiac conduction defects, iridescent posterior subcapsular cataracts, insulin-insensitive type 2 diabetes mellitus, and testicular failure.


There is no case report in the literature linking MD to MH. Lehmann-Horn and associates performed IVCT in 44 patients with myotonias and periodic paralyses, which revealed 4 positive, 10 equivocal, and 30 negative results.


Anesthetic Considerations


General considerations for MD are similar to those for other muscular dystrophies. Mathieu and coworkers conducted a retrospective study on the anesthetic and surgical complications of MD. The majority of complications were found to be pulmonary related and significantly more frequent in patients undergoing upper abdominal operations and those with severe disability, as assessed by the presence of proximal limb weakness. The pulmonary complications of MD are the result of hypotonia, chronic aspiration, and central and peripheral hypoventilation. Smooth muscle atrophy, which leads to poor gastric motility, when coupled with a diminished cough reflex, promotes aspiration.


Succinylcholine will produce contractions lasting for several minutes, thus making intubation and ventilation a challenge. These contractions are not antagonized by nondepolarizing muscle relaxants. Other agents, including methohexital, etomidate, propofol, and even neostigmine, may also induce myotonic reactions. Short-acting nondepolarizing muscle relaxants or avoidance of relaxation is therefore advised. Case reports have demonstrated normal responses to sugammadex when rocuronium was used as the neuromuscular blockade.


Triggering factors, such as hypothermia, shivering, and mechanical or electrical stimulation, may cause a myotonic reaction. The reaction can be treated with phenytoin (4-6 mg/kg/day) or quinine (0.3-1.5 g/day). Furthermore, MD patients can be very sensitive to anesthetic agents, with hypersomnolence and CO 2 retention sometimes being observed. Careful titration with relatively short-acting anesthetic agents may be beneficial. Close cardiac monitoring is required for MD patients. Pacing equipment should be readily available because a third of first-degree atrioventricular blocks may not respond to atropine. All patients should be treated as though they have both cardiomyopathy and conduction defects.




Myotonia Congenita


Myotonia congenita (MC) is a congenital form of muscular dystrophy characterized by uncontrolled temporary skeleton muscle excitability as a result of mutations in the muscle chloride channel gene ( CLCN1 ). There are two forms of MC, one with autosomal dominant and the other with recessive inheritance. The former is also known as Thomsen disease and the later as Becker myotonia. The myotonia in MC patients is usually initiated by a forceful muscle contraction, particularly after being at rest for at least 10 minutes. The myotonic muscle stiffness then becomes increasingly obvious after a second and third short, but forceful contraction. Further contractions usually dampen the myotonia.


Thomsen disease was the first myotonic disease to be described. Patients may have a hypertrophic and athletic appearance. The sign of percussion myotonia is described as an indenting-appearing myotonia triggered by tapping the muscle. Lid lag is common and muscle stretch reflexes are normal. Myotonia symptoms in Becker myotonia usually start at 10 to 14 years of age or even later and are more severe than those of Thomsen disease. Becker myotonia may be associated with severe generalized stiffness resulting in falling. It can frequently be misdiagnosed as epilepsy. Antiepileptic medications do improve the symptoms, however.


Anesthetic Considerations


As with many muscle diseases, there have been reports that MC patients are predisposed to MH, but as is the case for almost all of them, there are no data to support this hypothesis. However, depolarizing muscle relaxants can lead to severe masseter spasms in MC patients. Generalized spasms involving the respiratory and skeletal muscles have been reported. The findings resemble those of MH, so dantrolene is sometimes administered. Because dantrolene is an inhibitor of calcium release from the SR, it can usually abolish the myotonia effectively. Some believe that local anesthetics and class Ib antiarrhythmic drugs such as lidocaine should be used for myotonic reactions rather than dantrolene. Because shivering in the operating room can trigger the myotonic reaction, MC patients should be kept normothermic during surgery.




Myotubular Myopathy


Myotubular myopathy (MTM) is pathologically defined by the presence of centrally placed nuclei in the majority of extrafusal muscle fibers, an appearance resembling fetal myotubes during normal muscle development. Consequently, MTM is also called centronuclear myopathy (CNM). However, MTM now mostly refers to the X-linked form of the disease, whereas CNM is used for the autosomal form.


MTM and CNM are rare. The estimated incidence of MTM is 1 in 50,000 newborn males. MTM has been linked to the myotubularin ( MTM1 ) gene on Xq28. Pregnancy is often complicated by polyhydramnios and reduced fetal movements. Affected males typically have severe floppiness and weakness and respiratory distress at birth. Cardiac muscles are not generally involved. The patient usually has a normal response to pain, but tendon reflexes are absent. The long-term prognosis for MTM is extremely poor. In patients who survive the first year of life, most are either completely or partially ventilator dependent. These patients often have abnormal liver function. Both autosomal recessive and autosomal dominant forms have been observed in CNM patients. Its clinical features include respiratory distress, hypotonia, bulbar weakness, ophthalmoplegia, ptosis, and facial diplegia. Although the exact genetic mechanism is not fully understood, the MTM1 , myotubularin-related protein ( MTMR2 ), and myotubularin-related phosphatase ( MTMR3 ) genes have been implicated. Pathologically, MTM and CNM share a similar, characteristic histologic feature: predominantly type 1 fiber with centrally placed nuclei seen on hematoxylin-eosin staining of formalin-fixed, paraffin-embedded tissue.


Anesthetic Considerations


Reports of anesthesia for patients with MTM are sparse. Nontriggering general anesthesia has been used because of the unfounded concern of possible susceptibility to MH. Agents such as propofol, fentanyl, remifentanil, and nitrous oxide have been used successfully without adverse effects. The possibility of a prolonged effect of nondepolarizing muscle relaxants has been suggested with mechanomyography. However, in reality, intubation of such patients may not require any muscle relaxant because of their hypotonic state. Costi and van der Walt suspected that the defect in MTM is distal to the neuromuscular junction, whereas Dorchies and coworkers suggested that muscles in MTM might be intrinsically normal, with myotubularin-deficient motor neurons involved in development of the disease.




Metabolic Myopathies


Two major energy sources for muscle exist: glycogen and fatty acid. Glycogen serves as a dynamic, but limited reservoir of glucose, mainly stored in skeletal muscle and liver. Glycogen storage disorders (GSDs) are a group of metabolism disorders caused by enzyme deficiency or dysfunction. They reduce effective glucose storage by interfering with normal glycogen synthesis and degradation. Synthetic errors cause decreased normal glycogen, whereas degradation errors tend to block the breakdown of glycogen. Subsequently, hypoglycemia and accumulation of glycogen in tissues could occur as a result of substrate use. There are more than 12 types of GSD that are assigned roman numerals based on the enzyme deficiencies. Types I and II are discussed here.




Glycogen Storage Disease Type I


The incidence of GSD I is approximately 1 in 100,000 live newborns. The incidence in non-Ashkenazi Jews from North Africa may be as high as 1 in 5420 people. The defective enzyme causing the disease is glucose-6-phosphatase, which is the enzyme that converts glucose 6-phosphate (G6P) to glucose in the liver. Type Ia (von Gierke disease) is due to a deficiency in G6P hydrolase (catalytic subunit) activity and accounts for more than 80% of cases. Types Ib (G6P transporter deficiency), Ic, and Id represent allelic defects in the translocase associated with G6P. Their inheritance is autosomal recessive. The G6P gene ( G6PC ) encoding the hydrolase resides at 17q21, with the gene encoding G6P translocase ( G6PT ) located at 11q23. Mutations responsible for GSD I have been described in both type Ia and Ib patients.


Impaired glycogenolysis results in accumulation of glycogen and G6P in the liver, kidney, intestine, skeletal muscle, and heart and is manifested as hepatomegaly, renomegaly, proximal tubular dysfunction, and diarrhea. Fasting hypoglycemia is the initial manifestation of the disease. As a result, upregulation of the synthesis and transport of counter-regulatory hormones, such as glucagon, cortisol, catecholamines, and growth hormone, becomes obvious and leads to the release of pyruvate, lactate, and free fatty acid. Lipid deposition in lean tissues such as the liver, skeletal muscle, cardiac muscle, and pancreas results in lipotoxicity and organ failure, including pulmonary hypertension, steatohepatitis, end-stage renal disease, insulin resistance, cardiac contractile dysfunction, and pancreatic β cell failure. For type Ib disease, specific problems such as neutropenia and neutrophil dysfunction are prominent. Patients may have recurrent infections and inflammatory bowel disease.


Anesthesia Considerations


Anesthesia case reports for GSD I patients are rare. Patients with GSD I diseases should be given intravenous glucose-containing fluid preoperatively when they have fasted. Lactate-containing solutions should be avoided because these patients are not able to convert the lactic acid to glycogen. Patients need to be monitored frequently to avoid hypoglycemia.




Glycogen Storage Disease Type II (Acid Maltase Deficiency)


The incidence of acid maltase deficiency (AMD) is estimated to be 1 in every 14,000 to 40,000 births. Its inheritance is autosomal recessive with a few exceptions. Mutations of the acid maltase gene on chromosome 17q25 cause deficiency of lysosomal acid maltase (acid α-1,4-glucosidase). Cases of AMD have been arbitrarily classified into three groups—infantile, childhood, and adult—according to the age at onset or death, rate of progression, and tissue-organ involvement.


Acid maltase is a lysosomal enzyme that catalyzes the one-way hydrogenation of glycogen to G6P and is found in all tissues, including skeletal and cardiac muscle. Consequently, glycogen accumulates within the muscle tissues of maltase-deficient patients. Infantile AMD, also known as Pompe disease, is usually manifested within the first few months of life as rapidly progressive weakness and hypotonia and enlargement of the tongue, heart, and liver. Massive amounts of glycogen (8%-15% of the wet weight of the tissue) accumulate in the heart, liver, and skeletal muscle, with relatively smaller deposits in smooth muscle, eyes, kidneys, endothelial cells, lymphocytes, brain, and spinal cord. Accumulation of glycogen in cardiac muscle leads to cardiac failure in the infantile form. Echocardiography demonstrates marked thickening of the interventricular septum and posterior left ventricular wall, left ventricular outflow obstruction, and trabecular hypertrophy. Ventricular wall thickness may be increased to up to 25 mm. Wolff-Parkinson-White syndrome has been reported. The signs and symptoms of infantile AMD may resemble those of DMD. Death usually results from cardiorespiratory decompensation within several years of disease progression.


Childhood AMD occurs in infancy to early childhood and is manifested by clinical signs of myopathy. Respiratory muscles tend to be selectively affected. Calf enlargement can also occur. The disease progresses relatively slowly in this form, with a few patients surviving beyond the second decade of life. Tongue, heart, and liver enlargement occur infrequently. However, involvement of vascular smooth muscle is more severe than in the infantile form. There has been a report of extensive glycogen deposition in the arterial wall causing basilar aneurysms.


Adult AMD usually occurs after age 20 and is characterized by slow progressive myopathy or symptoms of respiratory failure. The weakness in proximal muscles is more prominent than the weakness in distal muscles. A third of adult AMD patients suffer from respiratory failure with a restrictive pattern. Weakness in the diaphragm causes extensive atelectasis. VC may be significantly reduced.


Anesthetic Considerations


Anesthesia reports in AMD patients are rare. Isolated intraoperative cardiac arrest during halothane anesthesia in infantile AMD has been documented. Despite the problem noted with halothane, enflurane and sevoflurane have been used without complications. Theoretically, total intravenous general anesthesia with propofol may cause a reduction in afterload leading to an increased risk for myocardial ischemia. This may become significant when the patient is tachycardic.


Subendocardial ischemia may occur in patients with a thickened ventricular wall and results in higher left ventricular end-diastolic pressure at lower ventricular volume. Close cardiac monitoring is therefore necessary. A central venous or pulmonary artery catheter is not essential in patients who are normovolemic without preexisting heart failure. Adequate filling pressure and normal to high systemic vascular resistance (SVR) are required to ensure effective coronary perfusion. Ketamine has been used successfully in a number of cases because of its ability to maintain SVR and contractility. Respiratory failure and muscle weakness are the other concerns for anesthesiologists. A spectrum of uses of muscle relaxants from none to atracurium to rocuronium have been attempted. Low-dose rocuronium, 0.5 mg/kg, with close monitoring of neuromuscular function and adequate use of reversal agents, has been sufficient to prevent prolonged postoperative weakness. Depolarizing agents should be avoided because of the potential risk of hyperkalemia and rhabdomyolysis.




Mitochondrial Myopathies


Mitochondrial diseases refer to defects in the five main steps of mitochondrial metabolism: substrate transport, substrate utilization, the Krebs cycle, the electron transport chain, and oxidation-phosphorylation coupling. However, the term mitochondrial myopathy has been reserved for disorders caused by defects in the respiratory chain. The respiratory chain is composed of five multimeric complexes (I-V) embedded in the inner mitochondrial membrane, plus two small mobile electron carriers, coenzyme Q 10 (CoQ 10 ) and cytochrome c , for a total of more than 80 proteins, among which 13 are encoded by mitochondrial DNA (mtDNA) and all others by nuclear DNA (nDNA). mtDNA is different from nDNA in several aspects: (1) mtDNA is circular and contains no intron, (2) it has larger numbers of copies than nDNA does and a much higher spontaneous mutation rate, and (3) its inheritance is maternal. Diagnosis of mitochondrial diseases is difficult because of their clinical heterogeneity.


Primary mtDNA mutations may include point mutations in polypeptide, tRNA, or rRNA encoding regions and large-scale rearrangements, duplications, or deletions. Some of the common conditions caused by point mutations include myoclonic epilepsy with ragged-red fibers (MERRF); mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS); neuropathy, ataxia, and retinitis pigmentosa (NARP); maternally inherited Leigh syndrome; and Leber hereditary optic neuropathy. Sporadic large-scale mutations may lead to Kearns-Sayre syndrome, progressive external ophthalmoplegia, and Pearson syndrome. nDNA mutations can cause deficiencies in complexes I to IV and CoQ 10 of the electron transport chain.


Mitochondrial diseases present a diagnostic challenge due to their clinical heterogeneity. Since mitochondria are ubiquitous; every tissue in the body can be affected by mtDNA mutations. Disorders due to nDNA mutations follow a Mendelian pattern and are thus phenotypically “all or none,” while inheritance of mtDNA is stochastic, leading to greater variability. Mitochondrial myopathy is estimated to have an incidence of 1 in 4000. Among all the mitochondrial functions, abnormalities in electron transport and oxidative phosphorylation are the most common causes of mitochondrial myopathies. Mitochondrial myopathies are characterized by proximal muscle weakness. Common laboratory findings include a very high lactate to pyruvate ratio (50-250:1 instead of a normal ratio of <25:1), increased blood levels of free carnitine, and occasionally low levels of folate (for instance in Kearns-Sayre syndrome). The hallmark of mitochondrial myopathies is the “ragged red fiber” when muscle biopsy specimens are stained with modified Gomori trichrome stain, and specific defects in the activity of these enzymes have been demonstrated in patients with mitochondrial disease. Fatigue and poor stamina are prominent clinical features. Movement disorders such as ataxia, dystonia, myoclonus, chorea, athetosis, and tremors have also been described as being due to mitochondrial abnormalities. CT and MRI scans of the brain may be very helpful—for example, patients with MELAS demonstrate basal ganglia calcifications and stroke-like patterns not confined to vascular territories. Clinical features of two relatively common encephalomyopathies, MELAS and MERRF, are briefly discussed below.


Mitochondrial Myopathy, Encephalopathy, Lactic Acidosis, and Stroke-Like Episodes


MELAS is the most common mitochondrial encephalomyopathy. Onset is most commonly before age 20. Seizures are common and stroke-like episodes (“stroke-like” because they do not conform to vascular distributions) may produce hemiparesis, hemianopia, and cortical blindness. Any patient with a stroke below the age of 40 should be worked up for MELAS. Associated findings include diabetes mellitus, hearing loss, pituitary and thyroid hypofunction, and lack of secondary sexual characteristics. In its full expression, MELAS leads to dementia, a bedridden state, and death. There is no specific treatment.


Myoclonic Epilepsy with Ragged Red Fibers


MERRF is a multisystem disorder characterized by myoclonus, generalized epilepsy, ataxia, and ragged red fibers on muscle biopsy. Other clinical features may include hearing loss, peripheral neuropathy, optic atrophy, dementia, short stature, and exercise intolerance. Cardiomyopathy is occasionally present. Laboratory features include an increased lactate at rest and exercise, and a myopathic picture on EMG and electroencephalogram showing generalized spike and wave discharges with background slowing. Only supportive treatment is available.


Anesthetic Considerations


The anesthesiologist may be involved in the care of patients with mitochondrial disease in multiple situations—often in the setting of obtaining a muscle biopsy in a child with an undiagnosed myopathy. These patients may also present for surgery for procedures related to the disease (such as implantation of a permanent pacemaker in a patient with KSS ), for incidental medical problems, as well as in the labor and delivery suite for labor analgesia. The diversity of clinical presentations encountered in the mitochondrial myopathies discourages a “one size fits all” approach to anesthesia. Rather, each patient should be thoroughly evaluated and the anesthetic plan tailored to the patient’s specific needs.


Preoperative Evaluation


Given the heterogeneous types of mitochondrial disease, patients with mitochondrial disease will need a comprehensive preoperative evaluation with a particular focus on neurologic, cardiac, respiratory, musculoskeletal, endocrinopathic, and metabolic compromise. An ECG and echocardiogram should be considered in patients with signs and symptoms of cardiomyopathy or conduction defects (or both). Although normal lactate and glucose levels cannot rule out mitochondrial diseases, laboratory studies consisting of glucose, electrolytes with anion gap, complete blood count, blood urea nitrogen, lactate, pyruvate, ammonia, CK, biotinidase, acyl carnitines, and blood and urine amino and organic acids could be used as initial investigation for suspected mitochondrial disorders. Further workup should include an erythrocyte sedimentation rate, glycosylated hemoglobin (HbA 1C ), liver and renal profiles, thyroid function tests, arterial blood gas, and urinalysis. Multidisciplinary consultation with special laboratory and imaging studies may be required.


Induction and Maintenance of Anesthesia


Anesthesia has a significant impact on mitochondrial function. Both barbiturates and propofol inhibit complex I of the electron transport chain. Local anesthetics have been demonstrated to disrupt oxidative phosphorylation and decrease the bioenergetic capacity of mitochondria. Sensitivity to intravenous barbiturates and etomidate has been reported. Fortunately, notwithstanding the potential pitfalls mentioned above, almost every anesthetic technique has been safely used in patients with mitochondrial disease. Midazolam, thiopental, propofol, remifentanil, and ketamine have all been used safely. Notably, propofol and midazolam are known to inhibit the mitochondrial respiratory chain in a dose-dependent manner. Indeed, mitochondrial dysfunction has been postulated as a mechanism for the propofol infusion syndrome. It is safe to use propofol as an anesthetic induction agent, but the use of a propofol infusion for long periods should probably be avoided.


Premedication should avoid respiratory depression caused by impaired respiratory responses to hypoxemia. Volatile agents such as halothane, isoflurane, and sevoflurane have been shown to inhibit complex I. This direct inhibition of mitochondrial electron transport system enzymes and altered mitochondrial bioenergetics in the heart were thought to be the mechanism of cardiac preconditioning by volatile anesthetics. Inhaled sevoflurane has been widely used for induction because of its low pungency. In some cases, halothane and isoflurane have also been used. Isoflurane has been recommended as the agent of choice in patients with Kearns-Sayre syndrome because rhythm disturbances have been reported with halothane in such patients. In addition, artificial pacing capability is recommended when dealing with this specific subset of patients. With use of the bispectral index, higher sensitivity to volatile agents has been suggested in children with mitochondrial diseases, especially with dysfunction of complex I. However, its methodology has been criticized. A decreased minimum alveolar concentration of halothane in patients with mental retardation has also been reported.


Despite no real evidence and the fact that volatile anesthetics are frequently the anesthetic of choice when muscle relaxants are considered, several papers have expressed concern that these myopathies are associated with increased sensitivity to MH. This conclusion is not supported by any data. Increased sensitivity to nondepolarizing muscle relaxants has been documented for mivacurium, atracurium, and rocuronium. In contrast, normal responses to depolarizing and nondepolarizing neuromuscular blockers such as pancuronium, vecuronium, and atracurium are also reported. Although muscle relaxants are not absolutely contraindicated based on current literature and research, it is necessary for anesthesia practitioners to cautiously administer depolarizing or nondepolarizing neuromuscular blockers to those patients with mitochondrial diseases and to use neuromuscular monitoring. Presently, there is no evidence to support an association between MH and mitochondrial disease. However, it may be prudent to avoid succinylcholine in patients with extensive myopathy to minimize the risk of hyperkalemia, although the safe use of succinylcholine has been documented in at least one patient with KSS.


Nonsteroidal antiinflammatory drugs and regional techniques consisting of local, spinal, and epidural administration have been reported. However, it is suggested that regional anesthesia be performed when neurologic abnormalities of the spinal cord and peripheral nerves have definitely been ruled out. Importantly, coagulation function should be assessed because of the possibility of hepatic dysfunction.


Opioids should be used with caution because of the increased risk for respiratory depression and their potential to induce respiratory acidosis in addition to the underlying metabolic acidosis. Because patients with mitochondrial diseases have dysfunctional aerobic metabolism, any increase in the basic metabolic rate should be prevented. Shivering, hypoxia, fasting, and hypotension in such patients may exacerbate the lactic acidosis and should therefore be avoided. Finally, the increased postoperative infection rate in patients with mitochondrial diseases may be due to low hepatic mitochondrial activity, phagocytosis by Kupffer cells, and decreased activity of the reticuloendothelial system.


Perhaps more important than the specific choice of anesthetic agents are the implications of patients’ co-morbidities and metabolic status. Normothermia should be maintained during surgeries, and intravenous fluids should be warmed to body temperature. Lactated Ringer (LR) should probably be avoided given the risk of preexisting lactic acidosis (although there is no evidence that LR has worsened acidosis when it has been used ). There have been multiple reports of hyponatremia (and occasional hyperkalemia) in these patients. Adrenal insufficiency should be considered in such a picture, particularly when accompanied by hypotension. Finally, these patients are at increased risk of cardiac conduction abnormalities and cardiomyopathy that should be taken into consideration while formulating an anesthetic plan ( Box 35.7 ).


Mar 7, 2020 | Posted by in ANESTHESIA | Comments Off on Neuromuscular Disorders Including Malignant Hyperthermia and Other Genetic Disorders

Full access? Get Clinical Tree

Get Clinical Tree app for offline access