Neurologic Diseases




Key points





  • Hereditary motor and sensory neuropathies (HMSNs) are disorders of myelination of the peripheral nervous system resulting in progressive loss of motor and sensory function.



  • In familial dysautonomia (HSAN-III), anesthesia management is directed at prevention and prompt treatment of dysautonomic crises.



  • Multiple system atrophy is a new term synonymous with Shy-Drager syndrome, striatonigral degeneration, and olivopontocerebrellar atrophy.



  • Parkinson’s disease symptoms include tremor, bradykinesia, rigidity, and postural instability; medical management restores dopaminergic versus cholinergic balance.



  • Huntington’s disease is characterized by choreoathetoid movements (butyrophenones and phenothiazines may temporarily alleviate), dementia, and psychiatric disturbances.



  • Sydenham’s chorea is an acute manifestation of rheumatic fever.



  • Dystonias are a broad group of movement disorders in which sustained involuntary muscle contractions lead to abnormal postures or repetitive movements.



  • Multiple sclerosis has an unpredictable pattern of relapse, which may coincide with postoperative recovery.



  • Guillain-Barré syndrome is characterized by ascending paralysis with areflexia.



  • Neurofibromatosis is a multisystem disease requiring a multidisciplinary surgical team for optimal outcomes.



  • Multisystem von Hippel–Lindau disease is characterized by CNS lesions combined with pheochromocytoma and renal cell carcinoma; most patients require neurosurgical procedures or surgery for removal of pheochromocytoma or renal cell carcinoma.



  • Tuberous sclerosis is also a multisystem disease, ideally requiring multidisciplinary team management; most patients require surgical procedures or imaging studies during childhood and typically have various degrees of mental retardation.



  • Sturge-Weber syndrome is a rare congenital (not heritable) vascular disorder of unknown etiology marked by facial angioma (port-wine stain) and leptomeningeal angioma.



  • Chiari malformation type I patients should be evaluated for degree of brainstem compression, cranial nerve involvement, spinal cord compression, and increased intracranial pressure.



  • In patients with Klippel-Feil syndrome, main consideration is catastrophic cervical spinal cord injury during induction of anesthesia and positioning.



  • Mucopolysaccharidoses can trigger new, potentially catastrophic neurologic complications; a difficult to impossible airway is the central concern.



  • Further anesthetic issues are detailed for specific neurologic diseases in the boxes.



Neurologic disorders represent a unique challenge to anesthesiologists by affecting the same biosubstrate targeted by anesthetic agents to produce the state of anesthesia. The direct consequence of neurologic disorders is often dramatically altered pharmacodynamics of anesthetics. Anesthetic agents are also in essence reversibly neurotoxic, and thus their use in patients with already-compromised neurologic function might lead to further deterioration of existing symptomatology. Monitoring the recovery of neurologic function from the effects of anesthesia is further complicated in patients with neurologic deficits. Many neurologic disorders also are very rare, with only minimal or no anesthesia experience reported in the literature.


This combination of factors in neurologic patients demands maximum caution and planning in the administration of anesthesia. The last two decades have seen rapid development in the understanding of genetic mechanisms and the underlying pathophysiology of many neurologic conditions, which in turn has led to better management of these disorders. It also helped to create newer, more precise classifications for many classes of neurologic disorders, guided more by etiology and pathophysiology than by pure symptomatology. However, significant overlap still exists between different classes of neurologic disorders, which is further complicated by cross-referencing to older eponymic classification. This chapter groups different disorders by the commonality of their anesthesia and perioperative challenges.




Hereditary peripheral neuropathies


Inherited disorders of peripheral nerves are a part of a much larger group of inherited or acquired polyneuropathies that often coexist with systemic, infectious, and metabolic diseases (e.g., diabetes mellitus, thyroid disease, neoplastic syndromes) or are caused by exposure to various agents (e.g., heavy metals, alcohol, certain medications). As such, the hereditary neuropathies are a common and diverse group of genetically determined neurologic diseases. They are primarily characterized by a dysfunction of peripheral sensory neurons in the presence of additional muscle weakness or autonomic system dysfunction. However, a dysfunction of the central nervous system (CNS) and other organ systems is more prominent in some types of hereditary neuropathies, which is of special relevance to the anesthesiologist treating these patients.


Historically, classification of the hereditary peripheral neuropathies was primarily based on clinical manifestations and eponyms were used to designate a specific combination of clinical symptoms (e.g., Riley-Day syndrome, Charcot-Marie-Tooth disease). However, significant phenotypic variability led to nosologic confusion. Modern classification of hereditary neuropathies is based on clinical and electrophysiologic characteristics, modes of inheritance, and underlying genetic mutations. The hereditary neuropathies are usually divided into three major groups according to their main clinical manifestation: predominantly motor involvement, predominantly sensory or autonomic involvement, or neither. These major groups are further divided into types (usually based on differences in clinical presentation, pathology, nerve conductivity studies) and subtypes (based on genetic characteristics). Table 8-1 summarizes some of the more prevalent hereditary types of polyneuropathy; a comprehensive description can be found in a recent review.



Table 8-1

Hereditary Peripheral Neuropathies *





















































HMSN/HSAN Type Clinical Manifestations/Underlying Pathology Inheritance Patterns Electrophysiologic Findings
Hereditary Primary Motor And Sensory Neuropathies (HMSNs)
HMSN type 1 (Charcot-Marie-Tooth disease type 1, CMT-1): five subtypes identified Demyelinating disorder, distal weakness, onset in 1st-2nd decade of life, slow progressing, “onion bulbs” Autosomal dominant Moderate to severe reduction in nerve conduction velocities
HMSN type 2 (Charcot-Marie-Tooth disease type 2, CMT 2): eight identified subtypes Neuroaxonal (not demyelinating) disorder, distal weakness, slow progressing Autosomal dominant Normal to mildly reduced nerve conduction velocities
HMSN type 3 (Dejerine-Scottas or congenital hypomyelinating syndrome): three identified subtypes Demyelinating disorder, severe hypotonia in early childhood or at birth, “onion bulbs” Autosomal dominant Severe reduction in nerve conduction velocities
HMSN type 4: seven identified subtypes Large group of disorders, typically with early, severe presentation and rapidly progressing, demyelinating, sometimes prominent sensory deficit Autosomal recessive Severe reduction or absent nerve conduction velocities
Hereditary Primary Sensory and Autonomic Neuropathies (HSANs)
HSAN type 1 (hereditary sensory radicular neuropathy) Small axon loss, acromutilation Autosomal dominant
HSAN type 2 (congenital sensory neuropathy) Large and small axon loss Autosomal recessive
HSAN type 3 (Riley-Day syndrome or familial dysautonomia) Large and small axon loss, with dysautonomic crises, lack lacrimation Autosomal recessive
HSAN type 4 (congenital insensitivity to pain with anhidrosis) Congenital sensory neuropathy with anhidrosis, C-axon loss (see text) Autosomal recessive

* Inherited neuropathies other than HMSN/HSAN include hereditary neuropathy with pressure palsy, hereditary brachial plexopathy, and giant axonal neuropathy.



As in the previous edition of this textbook, this chapter continues to use the modern classification, with cross-referencing to traditional eponyms. All major medical reference databases use this classification, and patients in clinical anesthesia practice increasingly are diagnosed according to this terminology, which is widely accepted in the mainstream neurologic practice.


Primary Hereditary Motor and Sensory Neuropathies, Including Charcot-Marie-Tooth Disease


The hereditary motor sensory neuropathies (HMSNs) represent a spectrum of disorders caused by a specific mutation in one of several myelin genes that results in defects in myelin structure, maintenance, and formation. The association of different mutations within the same gene with various clinical phenotypes is a common finding in this group of peripheral neuropathies. This variability suggests that these disorders represent a spectrum of related phenotypes caused by an underlying defect in peripheral nervous system myelination. The HMSNs, otherwise known as Charcot-Marie-Tooth disease (CMTD), have been classified as types 1 to 7, which are further subdivided, thus consisting of close to 30 clinical syndromes; the vast majority are very rare and have never been reported in the anesthesia literature. Because of the paucity of relevant anesthesia data on currently identified phenotypes, only Charcot-Marie-Tooth types 1 and 2 (CMT-1 and CMT-2) and HMSN type 3, which together are the most common hereditary peripheral neuropathies, are discussed here. Combined prevalence is almost 40 per 100,000 population.


Pathophysiology and Diagnosis


HMSN type 1, or CMT-1, is a demyelinating disorder of peripheral nerves that most often presents in the first or early-second decade of life, although infants can also be affected. Significant family history is typical. Diffuse slowing of nerve conduction velocity and gradually progressing distal muscle weakness with early loss of coordination characterize CMT-1. It is associated with loss of reflexes, talipes (pes) cavus, and hammer toe. Later, distal calf atrophy develops (classic “stork leg” deformity), in combination with gradual loss of proprioception and sense of vibration. Abnormal concentric myelin formations are called “onion bulbs” and are found around the peripheral axons. These are a characteristic feature of CMT-1, usually revealed by sural nerve biopsy. The CMT-1A subgroup of patients may present with proximal muscle wasting and weakness. Dematteis et al. also observed obstructive sleep apnea (OSA) in this subtype of patients, with a high degree of correlation between the severity of neuropathy and degree of obstruction. Even later changes include atrophy of the intrinsic hand and foot muscles, footdrop, palpable hypertrophy of the peripheral nerves, and possible scoliosis and kyphosis. Disease exacerbation may occur in pregnancy. Life expectancy is unaffected.


Type 2 HMSN, or CMT-2, also called axonal CMT, is a heterogeneous disorder with normal or borderline nerve conduction velocity. It is primarily an axonal, not a demyelinating, disorder, with neuropathy the result of neuronal death and wallerian degeneration (no onion bulbs on biopsy). The clinical course is similar to that of CMT-1, but sensory symptoms predominate over motor symptoms, and peripheral nerves are not palpable. Patients with CMT-2 C subtype display significant vocal cord and diaphragmatic weakness, resulting in OSA, which is of concern to the anesthesiologist. Onset is usually in the second or third decade of life, but can be seen in early childhood, with rapid clinical progression.


Type 3 HMSN includes two syndromes: Dejerine-Sottas syndrome and congenital hypomyelinating neuropathy (CHM). Both are characterized by profound hypotonia, presenting in early infancy or at birth, in the case of CHM. Dejerine-Sottas syndrome is clinically similar to CMT-1, although its manifestations are more severe and appear in early childhood.


The preoperative evaluation and preparation of HMSNs may be complicated because of similarity in clinical presentation with other genetic or acquired polyneuropathies ( Box 8-1 ).



Box 8-1

Differential Diagnosis of Hereditary Motor Sensory Neuropathies


Genetic Neuropathies





  • Refsum’s disease



  • Metachromatic leukodystrophy



  • Familial brachial plexus neuropathy



  • Adrenomyeloneuropathy



  • Pelizaeus-Merzbacher disease



  • Amyloid neuropathies



Acquired Neuropathies


Metabolic Disease





  • Diabetes mellitus



  • Thyroid disease



  • Vitamin B 12 deficiency



Infectious Disease





  • Neurosyphilis



  • Leprosy



  • Human immunodeficiency virus



Other Diseases





  • Chronic alcoholism



  • Heavy-metal intoxications



  • Vasculitis



  • Neoplastic syndromes



  • Chronic inflammatory demyelinating polyneuropathy




No specific treatments for HMSNs are available. Symptomatic supportive care consists of orthopedic corrective joint procedures for talipes cavus and scoliosis deformities and physical and occupational therapy. Orthopedic procedures are usually staged, ranging from soft tissue procedures and osteotomy to triple arthrodesis. Multiple administrations of general or regional anesthesia might be required.


Preoperative preparation of patients with HMSNs is dictated by the extent of clinical involvement and coexisting morbidities. The degree of motor neurologic involvement should be evaluated and affected muscle groups noted. Atrophic denervated muscles usually display significant resistance to nondepolarizing muscle relaxants and are unreliable for monitoring of neuromuscular blockade (NMB).


Patients with CMT-1 and CMT-2 C should be evaluated for restrictive pulmonary disease related to scoliosis and diaphragmatic weakness and potential OSA. Respiratory insufficiency has been described in patients with CMT. Careful planning for extubation and a possible need for postoperative respiratory support may be necessary in these patients. Patients with HMSNs may have undetected cardiac conduction abnormalities. Although this association is not strong, all patients with an HMSN should have a preoperative ECG. Pregnancy often leads to exacerbation of the symptoms of CMTD and, in combination with diaphragmatic splitting, can lead to respiratory compromise.


Anesthetic considerations


The anesthetic experience for HMSN types 1, 2, and 3 is limited to a number of case reports and retrospective reviews. Despite the absence of strong evidence advocating for or against the use of specific anesthetic agents or particular anesthetic techniques, a number of important concerns have been raised in the literature regarding anesthetic management of these patients ( Box 8-2 ).



Box 8-2

Hereditary Peripheral Neuropathies: Anesthetic Issues


Hereditary Motor and Sensory Neuropathies (HMSNs)





  • Informed consent regarding the effect of anesthetic agents on the course of the disease is difficult to provide because of a lack of conclusive evidence in the literature.



  • Thorough preoperative interview and maximal patient participation regarding the choice of anesthetic technique are advisable, combined with detailed documentation of these discussions.



  • Neuroaxial and regional anesthesia are not contraindicated, but lower concentrations of local anesthetic and careful titration are advisable.



  • Patients with advanced disease have increased risk of perioperative respiratory depression and sleep apnea.



  • Avoid monitoring neuromuscular blockade at the affected muscles to avoid overdose.



  • Use of depolarizing muscle relaxants should be avoided when possible in patients with HMSNs.



Hereditary Sensory and Autonomic Neuropathies: Familial Dysautonomia (HSAN-III)





  • Preoperative and intraoperative maintenance of euvolemia is critical to prevent severe hypotonic episode.



  • Invasive intraoperative hemodynamic monitoring is recommended, as dictated by degree of autonomic dysfunction and type of surgery.



  • Prolonged postoperative ventilatory support might be needed, especially in patients with compromised respiratory function.



  • Rapid-sequence induction should be considered in patients with a history of aspiration and gastroesophageal reflux.



  • Tight body temperature control is important the HSAN-III patient.




Drugs triggering malignant hyperthermia (MH) have been used in patients with CMTD without complication. However, in two reports of MH during general anesthesia in CMTD patients, the authors advocate against the use of succinylcholine and volatile agents. Furthermore, an approach postulating that any patient with a neuromuscular disease should be considered to be at increased risk for MH adds to this controversy. The review of the available literature describing anesthesia management in patients with HMSN indicates that most authors prefer to avoid administering MH-triggering agents in patients with HMSN types 1, 2, and 3, partly because of medicolegal considerations.


Succinylcholine use in these patients is associated with increased risk of malignant arrhythmias secondary to exaggerated hyperkalemic response. Although succinylcholine has been used in CMTD without untoward effects, it seems appropriate to avoid its use in any patient with suspected muscular denervation.


Nondepolarizing muscle relaxants have been used successfully in patients with HMSNs without indications of prolonged duration of action. However, some authors express reasonable concern that adequate monitoring of NMB could be complicated because of altered responses on the affected muscles, which are not always obvious during clinical assessment. Additionally, there is at least one case report of prolonged neuromuscular block with vecuronium. Inadequate reversal of NMB in patients with pre-existing respiratory compromise can lead to serious complications. Some advocate avoiding the use of nondepolarizing muscle relaxants in such patients whenever possible.


Neuroaxial anesthetic techniques have been successfully used in patients with HMSN without untoward effects, including for vaginal delivery and cesarean section. However, some authors correctly point out that medicolegal concerns must be considered when designing anesthesia plans for these patients. Despite lack of evidence that anesthesia affects the course of pre-existing neuromuscular disease, regional anesthesia may be erroneously blamed for any subsequent deterioration in sensory or motor deficits. This is especially true in pregnancy, which as noted, is associated with exacerbation of neurologic symptoms in women with CMTD. Additionally, the choice between general or regional/neuraxial anesthesia in patients with CMTD complicated by respiratory compromise is guided by the preservation of respiratory function in the perioperative period. In patients with phrenic nerve involvement, whose respiration depends on accessory muscles, regional block involving intercostal muscles can lead to acute respiratory failure. In other case reports, however, patients with CMTD required prolonged respiratory support after general anesthesia.


Patients with CMT-1 have demonstrated increased sensitivity to thiopental, correlating with the degree of motor and sensory deficit. However, propofol and total intravenous anesthesia (TIVA) have been successfully used in these patients without untoward effects.


Anesthetic management in the vast majority of patients with HMSN appears to be uncomplicated and should be directed to accommodate any coexisting systemic conditions. However, detailed preanesthesia assessment and thorough documentation of informed consent, with maximum patient participation in the decision-making process on choice of anesthesia technique, are crucial.


Hereditary Sensory and Autonomic Neuropathies


The hereditary sensory and autonomic neuropathies (HSANs) are a diverse and constantly expanding group of disorders affecting the development of autonomic and sensory neurons. Until recently, seven such disorders have been described, with familial dysautonomia (HSAN type III), also known as Riley-Day syndrome, and HSAN type IV, also known as congenital insensitivity to pain with anhidrosis (CIPA), the most recognized and well understood. All HSANs are manifested by both sensory and autonomic dysfunction present at variable degrees, with a unique feature for all types being absence of a normal axon flare response after intradermal injection of histamine phosphate. The reported anesthetic experience for HSANs is limited to anesthesia management of patients with familial dysautonomia (HSAN-III) and CIPA (HSAN-IV). The clinical presentation, diagnosis, and management of other HSANs have been reviewed.


Familial dysautonomia (HSAN Type III, or riley-day syndrome)


Familial dysautonomia (FD) is a rare genetic disorder that affects, almost exclusively, persons of Ashkenazi Jewish extraction. It is the most prevalent and well studied of all HSANs. Development of autonomic and sensory neurons is impaired, resulting in reduced population of nonmyelinated and small-diameter myelinated axons. The sympathetic neurons are primarily affected, and sympathetic ganglia are small. The parasympathetic neurons and large axons are generally spared. FD presents at birth and progresses with age. In the past, more than 50% of patients died before 5 years of age. Currently, because of improvements in diagnosis and treatment, newborns diagnosed with FD have a greater than 50% chance to live past age 30.


Although FD has close to 100% penetrance, the presentation of the disease at different life stages is highly variable. Autonomic dysfunction is the most prominent feature, presenting the greatest impediment to normal functioning, and usually overshadows the sensory deficits. Dysautonomic crisis is the most dramatic feature of this syndrome, characterized by episodes of severe nausea and vomiting associated with agitation, hypertension, tachycardia, excessive sweating, and salivation, which are easily triggered by emotional or physical stress, or arousal from sleep.


The clinical diagnosis of FD is usually established soon after birth by demonstrating the presence of four main criteria: absence of tears with emotional crying, absence of lingual fungiform papillae, hypotonic or absent patellar reflexes, and absence of axon flare to intradermal injection of histamine in children of Ashkenazi Jewish descent. Many other systems are affected at various stages in life, and the myriad of clinical manifestations of FD can be divided into two main groups: sensory dysfunction and autonomic dysfunction ( Box 8-3 ). Differential diagnosis typically does not present a problem, considering availability of genetic testing and the fact that this disease is restricted to Ashkenazi Jews. However, many other conditions have some similar symptoms of autonomic and sensory dysfunction. All HSANs, cranial nerve and nuclear dysplasias, cri du chat (cat’s cry) syndrome, and Möbius syndrome can have some of the features found in FD. Many eye conditions have similar ocular manifestations with those of FD.



Box 8-3

Familial Dysautonomias: Sensory and Autonomic Dysfunction


Sensory System





  • Decreased pain sensation, often with hypersensitivity of palms, sole, neck, and genital areas; decreased temperature sensation.



  • Visceral sensation is intact.



  • Sense of vibration and proprioception is affected in older individuals; ataxia.



  • Hypotonia in younger children; often disappears with age; decreased tendon reflexes.



  • Prone to self-injury; unrecognized fractures; scoliosis and joint deformities.



Autonomic System


Gastrointestinal System





  • Impaired oropharyngeal coordination and impaired swallowing, resulting in dysphagia and frequent aspirations in newborns and infants.



  • Abnormal esophageal motility; decreased lower esophageal sphincter pressure and esophageal reflux.



  • Gastrointestinal dysmotility, complicated by cyclic vomiting (part of dysautonomic crisis).



Respiratory System





  • Recurrent pneumonias result from aspirations.



  • Insensitivity to hypoxia and hypercapnia (no ventilatory response).



  • Low tolerance for hypoxia; profound hypotension and bradycardia in response to hypoxia.



Cardiovascular System





  • Rapid, severe orthostatic hypotension without compensatory tachycardia.



  • Episodes of severe hypertension and tachycardia as part of dysautonomic crisis.



  • Syncopal episodes produced by various stimuli (e.g., full bladder, large bowel movement).



  • Postural hypotension.



  • Postural hypertension can develop in older patients.



Dysautonomic Crisis





  • Episodes of severe nausea and vomiting associated with agitation, hypertension, tachycardia, and excessive sweating/salivation.



  • Easily triggered by emotional or physical stress and arousal from sleep.



Other Manifestations


Renal System





  • Dehydration azotemia.



  • Progressive loss of renal function with age.



Central Nervous System/Developmental





  • Emotional lability, probably related to catecholamine imbalance.



  • Prolonged breath holding with crying, decerebrate posturing, syncope, cyanosis; may be misinterpreted as seizures.



  • Normal intelligence.



  • Delayed development.



Ocular Manifestations





  • Absence of overflow tears with emotional crying in all patients.



  • Corneal insensitivity; abrasions and spontaneous injuries; ulcers.



  • Optic neuropathy increases with age.



Laboratory Findings





  • Elevated blood urea nitrogen (BUN).



  • Hyponatremia associated with excessive sweating.



  • Catecholamine imbalance: elevated dopa/DHPG ratio.



Dopa, 3,4-Dihydroxyphenylalanine; DHPG, 3,4-Dihydroxyphenylglycol.



Treatment of FD patients is symptomatic. Diazepam is the most effective treatment for dysautonomic crisis with vomiting. It also normalizes blood pressure and heart rate in these patients. Increased salt and fluid intake is used to treat dehydration and hyponatremia and associated postural hypotension. Fludrocortisone and midodrine are also used for this purpose. Surgical procedures performed on these patients include gastrostomy in the majority of patients before age 5 years, to provide fluid and alimentation in patients with dysphagia; fundoplication for treatment of gastroesophageal reflux (GER) and associated pneumonia; and spinal fusions for severe scoliosis.


Anesthetic considerations


Anesthesia for surgical procedures had been associated with great risks in patients with FD. Recent progress in the understanding of existing risks and improved preoperative preparation resulted in significantly improved perioperative outcomes. Good working knowledge of FD manifestations and a systematic approach to preoperative assessment are essential for successful anesthetic management of these patients (see Box 8-2 ).


The respiratory system should be evaluated for signs of chronic or acute infections from repeated aspirations. Chest radiography is warranted in all patients. In patients with restrictive pulmonary disease caused by chronic pneumonias and scoliosis, arterial blood gas (ABG) analysis is included.


Severe intraoperative hypotonia is a well-recognized risk of general anesthesia. Cardiac output depends on preload because of a lack of compensatory sympathetic response to hypotonia. Correction of existing dehydration and hyponatremia is essential for intraoperative hemodynamic stability in these patients. Intravenous (IV) prehydration with crystalloids is often recommended to achieve euvolemic status preoperatively. Patients are evaluated for the presence and severity of GER; antacids need to be administered preoperatively to affected patients. Renal function is assessed to rule out significant renal failure, which can affect the choice of muscle relaxants. Patients with FD are prone to anticipation anxiety that can trigger dysautonomic crisis. Preoperative medication with benzodiazepines is recommended. Preoperative medication with opioids is contraindicated because of possible increased sensitivity to the agents.


Intraoperative management of FD patients is directed toward better cardiovascular stability, prevention of pulmonary aspiration, prevention of postoperative respiratory compromise, and adequate postoperative pain control. Invasive hemodynamic monitoring (intra-arterial line and central venous catheters) has been advocated in the past but was not used in one reported series without untoward effects. It appears reasonable to use invasive monitoring in patients with postural hypotension and for extensive surgery with large fluid shifts. Immediate preinduction administration of fluid bolus can reduce blood pressure (BP) variation. BP instability intraoperatively is treated by additional fluid boluses and direct-acting vasopressors, if the patient is unresponsive to administration of fluids. Any episodes of desaturation are promptly addressed by increased oxygen concentration to avoid profound hypotension and bradycardia from lack of hypoxic compensatory responses.


Rapid-sequence induction with cricoid pressure should be considered in patients with GER and a history of repeated aspirations.


Careful planning for extubation, postoperative ventilatory support, and weaning from the respirator in the intensive care unit (ICU) should be part of the routine postoperative management for these patients. In the past, FD patients frequently required prolonged ventilation in the ICU setting after general anesthesia. Reports indicate that with alternative techniques, such as epidural or local anesthesia, or deep propofol sedation with spontaneous ventilation, these patients can recover from anesthesia quickly without need for postoperative respiratory support.


Although patients with FD have decreased perception of pain and temperature, their visceral perception is intact, and they need sufficient levels of anesthesia and postoperative pain control. Postoperative pain should be promptly treated to avoid dysautonomic crisis. Nonsteroidal anti-inflammatory drugs (NSAIDs) or paracetamol will suffice in many cases. Opioids should be used cautiously to avoid respiratory depression. Regional techniques can be useful.


There have been no reports of adverse or prolonged responses of FD patients to any specific anesthetic agents or muscle relaxants. For appropriate surgical procedures, regional anesthesia is well tolerated. Use of deep propofol sedation for endoscopic outpatient procedures has been reported, with excellent results. Body temperature needs to be carefully monitored because of impaired temperature control in these patients. The eyes should be lubricated and protected at all times.


Familial dysautonomia is a serious anesthetic challenge that can be hazardous in these patients without proper preoperative preparations and intraoperative management. However, current approaches have resulted in significantly reduced mortality and morbidity in FD patients.


Congenital insensitivity to pain with anhidrosis


Pathophysiology and Diagnosis


Congenital insensitivity to pain with anhidrosis, or HSAN type IV, is a rare autosomal recessive neuropathy characterized by recurrent episodic fever, anhidrosis (absence of sweating), pain insensitivity, self-mutilating behavior, and mental retardation. Death from hyperthermia has been reported in infants with CIPA. Besides anhidrosis, it differs from FD by complete insensitivity to superficial and deep painful stimuli and normal lacrimation, much milder autonomic dysfunction, with absent postural hypotension or dysphagia. Self-inflicted multiple injuries are typical for these patients. This is often accompanied by accidental trauma, burns, wound infections, skin ulcers, joint deformities, and osteomyelitis.


There is only limited anesthetic experience in patients with CIPA. Okuda et al. suggest three important considerations in the anesthesia management of patients with CIPA: anxiety alleviation, temperature control, and adequate pain control. Despite congenital insensitivity to pain, general anesthesia was found to be necessary. Overall requirements of general anesthetics necessary for maintaining stable hemodynamics have been only slightly reduced. General anesthesia was used in all patients in these reports without any adverse reactions to the IV or inhalational anesthetic agents, opioids, or succinylcholine. In one report, a patient died after intraoperative cardiac arrest without clear cause, although the authors suspected that the high concentration of halothane used (2%) could have been responsible. Previous recommendations against the use of atropine (or other anticholinergic drugs) to avoid hyperpyrexia in these patients was not supported by the results reported in this series. Many patients received atropine without untoward effects.




Neurodegenerative disorders with autonomic failure


Autonomic failure (or dysautonomia), with its protean range of manifestations and symptoms, is a common part of an immensely diverse group of disorders in which some or all elements of the autonomic nervous system are affected. Autonomic failure to varying degrees is a part of the presentation of many systemic diseases (e.g., diabetes mellitus, amyloidosis), infectious diseases (e.g., leprosy, human immunodeficiency virus [HIV], rabies), immune disorders (e.g., acute dysautonomia, Guillain-Barré syndrome), paraneoplastic disorders, hereditary autonomic disorders (e.g., all HSANs, dopamine β-hydroxylase deficiency), and neurodegenerative disorders. A comprehensive discussion on various aspects of autonomic dysfunction in these conditions can be found in most neurology and medical textbooks. This section discusses only the most prevalent neurodegenerative disorders in which autonomic failure plays a prominent role, presenting a significant anesthetic challenge.


Parkinson’s disease (PD), dementia with Lewy body disorder (LBD), multiple system atrophy (MSA), and pure autonomic failure disorder (PAF) are all neurodegenerative disorders of unclear etiology, presenting with variable degrees of autonomic dysfunction. Based on the differences in the neuropathology, these disorders can be divided into two subgroups: Lewy body syndromes (PD, LBD, and PAF) and multiple system atrophy. All these disorders are characterized by the presence of α- synuclein in the neuronal cytoplasmic inclusions (Lewy bodies, as in Lewy body syndromes) or the glial cell inclusions (GCIs, as in MSA); thus these disorders are often called synucleinopathies. In PD, neurodegeneration is predominant in the substantia nigra and other brainstem nuclei and in peripheral autonomic neurons. Motor dysfunction is more prominent than autonomic failure in PD patients. Neuronal degeneration in PAF is restricted to peripheral autonomic neurons; thus the symptoms of pure autonomic failure without other manifestations. Extensive cortical involvement, in addition to degeneration of brainstem nuclei and peripheral autonomic neurons, is characteristic for LBD, which presents as severe dementia associated with parkinsonism and autonomic failure.


In MSA, cytoplasmic inclusions are found in the glial cells (GCIs) and not neurons (Lewy body). These inclusions are associated with degenerative changes in the central neurons in basal ganglia, cortex, and spinal cord, but not in peripheral autonomic neurons. Two phenotypes of MSA are currently identified based on the predominant clinical picture of parkinsonism (MSA-P) or cerebellar dysfunction (MSA-C). In the past, the patients with a predominant picture of autonomic failure were diagnosed with Shy-Drager syndrome. Currently, this term is rarely used, because all patients with MSA have a significant degree of autonomic dysfunction.


Autonomic failure in patients with Lewy body syndromes and MSA typically manifests with orthostatic and postprandial hypotension, bladder dysfunction, gastrointestinal (GI) motility disorders, and erectile dysfunction (ED). Orthostatic and postprandial hypotension is often the earliest and most disabling aspect of dysautonomia in many patients. Other symptoms of autonomic dysfunction, as described for familial dysautonomia, can be present. The differential diagnosis can be difficult because of frequent overlapping of the clinical picture between these conditions, especially in the initial stages of the disease process. Definitive diagnosis in some disorders could be established only on postmortem histopathologic examination. However, thorough clinical examination helps to distinguish between PD, LBD, MSA, and PAF ( Table 8-2 ). The subject of neurodegenerative disorders with autonomic failure has been reviewed.



Table 8-2

Differential Diagnosis of Multisystem Atrophy, Parkinson’s Disease, Pure Autonomic Failure, and Dementia with Lewy Bodies

Modified from Marti MJ, Tolosa E, Campdelacreu J: Mov Disord 18(suppl 6):21-27, 2003; and Kaufmann H, Biaggioni I: Semin Neurol 23:351-363, 2003.












































































Characteristic Multisystem Atrophy Parkinson’s Disease Pure Autonomic Failures Dementia with Lewy Bodies
Central nervous system involvement Multiple involvements Multiple involvements Unaffected Multiple involvements
Site of lesions Mainly preganglionic, central; degeneration of interomediolateral cell columns Peripheral autonomic postganglionic neurons Mainly peripheral autonomic postganglionic neurons; loss of ganglionic neurons Cortex, brain stem, peripheral autonomic postganglionic neurons
Progression Fast; median survival, 6-8 years after first symptoms Slow Slow; up to 15 years and longer Slow
Prognosis Poor Good Good Moderate to poor
Autonomic dysfunction Early onset, severe Late onset, usually mild to moderate Severe, usually the only manifestation Unclear, but can be severe
Extrapyramidal involvement Common Common Absent Common
Cerebellar involvement Common Common Absent Common
Lewy bodies Mostly absent Primarily in substantia nigra Present in autonomic neurons Cortex, brainstem, hippocampus
Glial cytoplasmic inclusions (postmortem staining) Present Absent Absent Absent
Response to chronic levodopa therapy Poor Good Moderate
Dementia Uncommon Usually not severe, 25%-30% of patients Uncommon Early, severe, rapidly progressing


Anesthetic management of PD is described later. Although LBD is the second most common cause of dementia after Alzheimer’s disease, there are no reports of anesthetic management in the literature. It appears reasonable to assume that the principles of anesthetic management of patients with LBD are common to those in patients with other forms of dementia. In LBD patients with advanced dysautonomia, the same precautions should be taken as in patients with MSA.


Multiple System Atrophy


In 1998, consensus committees representing the American Autonomic Society and the American Academy of Neurology defined multiple system atrophy as a sporadic, progressive, neurodegenerative disorder of undetermined etiology, characterized by features in the three clinical domains of parkinsonism, autonomic failure, and cerebellar or pyramidal dysfunction. In the past, the terms striatonigral degeneration, olivopontocerebellar atrophy, and Shy-Drager syndrome were used, depending on the predominance of clinical symptoms in any of these three domains.


Multiple system atrophy is a fatal disease that typically presents in the fourth to sixth decade of life, with mean disease duration of 6 years from onset of symptoms. Because of the significant similarity of clinical presentation to other neurodegenerative disorders, MSA is often not diagnosed until later stages. Parkinsonism is a predominant symptom in 80%, and cerebellar dysfunction in 20%, of all patients. Parkinsonism is usually not responsive to antiparkinsonian medications, which helps to differentiate MSA from PD. The most common and early presentation of autonomic dysfunction is urinary incontinence and ED. Orthostatic hypotension is found in half of MSA patients and is usually mild. Reduced heart rate variability and absence of compensatory tachycardia during hypotension is characteristic. Paradoxically, supine hypertension is present in more than half of patients and complicates their management. Recurrent syncope is a sign of severe orthostatic hypotension. Severe constipation, fecal incontinence, and decreased sweating are other signs of autonomic dysfunction in MSA.


Obstructive sleep apnea or central sleep apnea and sleep-related inspiratory stridor associated with bilateral vocal cord paresis or dysfunction have been reported in MSA patients.


There are no currently available treatments that can modify the clinical course or address the underlying pathologic MSA process. All the treatments are symptomatic, intended for improving the quality of life in these patients. Orthostatic hypotension is treated with administration of fludrocortisone or milrinone (oral adrenergic vasoconstrictor). The presence of significant supine hypertension limits the use of vasopressors. Erythropoietin has been reported to be useful in the treatment of patients with associated anemia and severe hypotension. Tracheostomy and respiratory support is reserved for the patients with stridor and central sleep apnea.


Anesthetic considerations


Perioperative management of patients with MSA is a formidable challenge because of potential hemodynamic instability and respiratory compromise in the postoperative period ( Box 8-4 ). A few case reports in the literature indicate no adverse effects to most of the common anesthetic agents. The management is directed at ensuring hemodynamic stability through invasive hemodynamic monitoring, adequate preoperative hydration, and maintenance of normovolemia with fluid replacement intraoperatively. Preoperative optimization of fludrocortisone therapy is recommended. Some controversy surrounds the potentially unpredictable response to vasopressor amines because of sympathetic hypersensitivity caused by autonomic denervation. Therefore, it is recommended to administer vasoactive medications very cautiously in much smaller doses than usual. However, vasopressors have been used without adverse effects for treatment of hypotension intraoperatively, when titrated judiciously.



Box 8-4

Neurodegenerative Disorders with Autonomic Failure: Anesthetic Issues


Multiple System Atrophy (MSA)





  • Perioperative hemodynamic instability, related to autonomic failure with orthostatic hypotension and sometimes severe supine hypertension, is to be expected. Invasive hemodynamic monitoring may be of benefit in perioperative management of patients with severe symptoms.



  • Obstructive and central sleep apnea and sleep-related inspiratory stridor may occur. The risk of postoperative airway obstruction and apneic episodes is increased in patients with MSA.



  • In patients with advanced dementia and parkinsonism, anesthetic considerations are similar to those in patients with other forms of dementia and Parkinson’s disease.



  • No specific anesthetic techniques or agents are contraindicated in MSA patients.



Pure Autonomic Failure





  • Anesthetic considerations are similar to those in patients with MSA.




Significant intraoperative supine hypertension has been reported, with minimal response to labetalol but profound hypotension after hydralazine administration. The hypotension responded only to vasopressin infusion. Short-acting vasodilators such as sodium nitroprusside may be a better choice for intraoperative supine hypertension. The hypertensive episodes in autonomic failure are particularly responsive to transdermal nitroglycerin.


Neuraxial anesthesia techniques have been successfully employed in patients with MSA, including for labor and delivery, with a greater degree of hemodynamic stability, also avoiding possible difficulties with extubation in these patients. It is speculated that patients with autonomic failure are less likely to respond with hypotension to sympathectomy caused by neuraxial block because they are already sympathectomized. The data in the literature support this hypothesis.


When general anesthesia is chosen, careful planning for extubation and postoperative monitoring of the respiration in the ICU setting is warranted, especially in MSA patients with a history of stridor or central or obstructive sleep apnea.


Pure Autonomic Failure


Pure autonomic failure is a sporadic, slow-progressing neurodegenerative disorder of the autonomic nervous system (ANS) that typically affects individuals in the sixth decade of life. PAF is characterized by an isolated impairment of the peripheral and central ANS. No symptoms of parkinsonism, cerebellar dysfunction, or dementia are usually present. The orthostatic hypotension in this syndrome is typically severe and more disabling than in other neurodegenerative disorders with autonomic failure. Other symptoms of autonomic failure are similar to those seen in MSA. The prognosis in PAF patients, however, is much better.


There is only one case report in the literature of general anesthesia without complications in a patient with PAF; it is unclear whether the patient also had epidural anesthesia performed. However, the authors advocate the use of epidural anesthesia and invasive hemodynamic monitoring for greater hemodynamic stability.


The same principles of anesthetic management used for patients with MSA should be applied when managing PAF patients.




Basal ganglia and cerebellar disorders


Parkinson’s Disease


Parkinson’s disease is a chronic progressive neurodegenerative disease characterized by resting tremor, bradykinesia, rigidity, and postural instability. In addition to these cardinal signs, many patients with PD will experience secondary symptoms attributable to the disease or its pharmacotherapy. Dementia is common, especially in advanced stages, as are fatigue and depression. Hallucinations, psychosis, anosmia, and autonomic instability are also well-described nonmotor features of the disease. Diagnosis is clinical, requiring at least two of the four cardinal signs. Prevalence has been estimated at approximately 1% of the population over 60. To date, attempts to identify risk factors have produced contradictory results. Older age has been persistently associated with increased risk of PD. Other putative risk factors include environmental exposure to heavy metals, pesticides, and herbicides; dietary factors; and body weight. Although most cases appear sporadic, several genes have recently been linked to PD, particularly in those cases manifesting before age 50.


Pathophysiology


Coordination of movement depends on a complex feedback loop in which the cortex sends information to the basal ganglia and cerebellum and in turn receives information from these structures through the thalamus. Because of their anatomic location, these pathways are often referred to as “extrapyramidal.” PD is characterized by neuronal loss, depigmentation, and gliosis in the substantia nigra pars compacta and pontine locus ceruleus, as well as degeneration of the putamen, globus pallidus, hippocampus, and brainstem nuclei. The result of this degeneration is a relative dopamine deficiency, particularly in the striatum and putamen, and unopposed cholinergic activation of inhibitory γ–aminobutyric acid (GABA) transmission from the striatum. The end result of this imbalance is excessive inhibition of the thalamus, which suppresses both the cortical motor system, leading to bradykinesia and tremor, and the brainstem motor areas, leading to gait and postural instability.


The precise mechanism of degeneration in PD has yet to be elucidated. Programmed cell death, protein misfolding and aggregation, abnormal proteosomal degradation, oxidative stress, mitochondrial dysfunction, and immunomodulation and inflammation have all been proposed. Although no universally accepted pathologic criteria exist for the diagnosis of PD, one hallmark of the disease appears to be the presence of eosinophilic, intracytoplasmic inclusions known as Lewy bodies. Composed primarily of α-synuclein, in association with ubiquitin, complement, and numerous cytoskeletal proteins, Lewy bodies are found in the substantia nigra, locus ceruleus, cerebral cortex, and sympathetic ganglia of PD patients, as well as in the cardiac sympathetic plexus, dorsal vagal nucleus, and myenteric plexus of the intestines. Importantly, Lewy bodies are not specific for PD and are found in other neurodegenerative diseases (e.g., MSA, progressive supranuclear palsy, Lewy body dementia), in Down syndrome, in amyloidopathies (e.g., Alzheimer’s disease), in tau -protein–associated diseases (e.g., frontotemporal dementia), and even in a small percentage of normal elderly brains. It remains unclear whether Lewy bodies represent a pathologic neurotoxic process, or as recent evidence suggests, play a neuroprotective role.


Medical Management


Pharmacologic management of PD patients seeks to balance dopaminergic and cholinergic effects in the striatum and thus preserve motor function and quality of life while minimizing medication-related side effects. Current therapies attempt to achieve these goals by blocking acetylcholine transmission, enhancing dopamine production, or stimulating central dopamine receptors ( Table 8-3 ). Motor symptoms generally respond better to treatment than extramotor symptoms, but effectiveness diminishes in later stages. Although several agents have shown possible neuroprotective effects in human and animal studies, currently no treatment significantly reverses or delays PD progression.



Table 8-3

Pharmacologic Therapy of Parkinson’s Disease




































































Drug Features Side Effects Contraindications
Dopamine Precursors
Levodopa Converted to dopamine by dopa decarboxylase
Combined with carbidopa (see Sinemet)
Treats akinetic symptoms, tremor, and rigidity
Not as effective for postural instability
Motor fluctuations and dyskinesias often become impediments to therapy.
Evidence for both neurotoxic and neuroprotective effects
Nausea, somnolence, dizziness, headache, confusion
Hallucinations/delusions
Agitation
Pyschosis
Motor fluctuations (“wearing-off phenomenon”)
Dyskinesias/dystonias
Homocysteine elevation (proposed)
Peripheral neuropathy if concomitant elevated methylmalonic acid
History of psychosis
Narrow-angle glaucoma
Concurrent monoamine oxidase inhibitor (MAOI) therapy
Carbidopa Decarboxylase inhibitor that does not cross blood-brain barrier
Prevents peripheral conversion of levodopa to dopamine
Reduces nausea, vomiting, orthostatic hypotension
Rebound hypertension with withdrawal
Sinemet Combination of carbidopa/levodopa
Available in 1:10 and 1:4 ratio
Controlled-release formulation also available
Dopamine Agonists
Bromocriptine
Cabergoline
Pramipexole
Ropinirole
Apomorphine
Sometimes used as levodopa-sparing monotherapy in younger patients
Fewer “on-off” fluctuations and less dyskinesia than levodopa
Apomorphine used as parenteral “rescue” therapy for levodopa-induced motor fluctuations
Similar to levodopa
Dopaminergic dysregulation syndrome (compulsive use, cyclic mood disorder)
Impulse control disorders
Dopamine withdrawal syndrome (resembling cocaine withdrawal)
Valvular heart disease (cabergoline)
Angina and orthostasis (apomorphine)
Acute somnolence (pramipexole)
History of psychosis
Recent myocardial infarction
Severe vascular disease
Breastfeeding
Anticholinergics
Trihexylphenidyl Benztropine Most useful in patients under age 70 without significant akinesia or gait disturbance
May be used adjunctively for persistent tremor
Improve DA/Ach balance
Rapid withdrawal may precipitate exacerbation of symptoms
Common and may limit use
Memory impairment, confusion, hallucinations
Peripheral antimuscarinic effects
Relatively contraindicated in elderly or cognitively impaired patients
Prostatic hypertrophy
Closed-angle glaucoma
Catechol O -Methyltransferase (COMT) Inhibitors
Tolcapone
Entacapone
Prolong and potentiate levodopa effect by increasing plasma half-life Dyskinesia
Hallucinations
Confusion
Nausea
Orthostatic hypotension
Hepatotoxicity (tolcapone)
Diarrhea
Liver disease may be a relative contraindication.
Liver function must be monitored for the first 6 months in all patients.
Monoamine Oxidase B (MAO-B) Inhibitors
Selegiline
Rasagiline
Modest effect in some patients
Inhibit breakdown of DA
May be neuroprotective
Nausea
Headache
Diarrhea
Insomnia (selegiline’s amphetamine metabolites)
Confusion in elderly
Use caution with concomitant SSRIs or TCAs
Does not precipitate hypertension with concomitant tyramine ingestion
Antivirals
Amantadine Mild antiparkinsonian activity
Mechanism unclear (likely dopaminergic, anticholinergic, and NMDA mediated)
Short-term monotherapy for mild disease
May reduce levodopa-induced dyskinesia and motor fluctuations
May reduce impulse control disorders in patients taking dopamine agonists
Side effects are rare in monotherapy but include livedo reticularis, ankle edema, hallucinations, confusion, and nightmares.
Central nervous system side effects are more likely when used adjunctively.
Hormone Replacement
Estrogen Adjunctive therapy in postmenopausal women
Effect may be indirect through improved subjective well-being.
Improved “on time,” but not objective scales of ADLs in one study
Few data on estrogen/progesterone
Adverse effects associated with long-term estrogen use

DA, Dopamine; Ach, acetylcholine; SSRIs, selective serotonin reuptake inhibitors; TCAs, tricyclic antidepressants; ADLs, activities of daily living.


Surgical Management


Surgical treatment of PD began in the early 20th century and was originally based on the intentional lesioning of deep brain structures. Thalamotomy was performed to ameliorate tremor and pallidotomy to control both parkinsonism and levodopa-induced dyskinesias. Although the introduction of stereotactic localization improved outcomes and reduced complications from these surgeries, the continued risk of permanent paresis, visual field cuts, gait disturbances, dysarthria, and hypersalivation, combined with the development of deep-brain stimulation techniques, have led to a pronounced shift away from permanent surgical lesioning.


Drawing on the intraoperative observation that focal electric stimulation of the brain induced a functional but reversible “lesion,” implantable deep-brain stimulation of the subthalamic nucleus (STN), globus pallidus (GPi), and ventralis intermedius (Vim) have all been successful in ameliorating PD symptoms. Vim stimulation appears to be most effective for tremor-predominant cases. Evidence from randomized controlled trials (RCTs) suggests that both STN and GPi are effective for treatment of motor symptoms and bradykinesia. STN stimulation appears to provide a greater dose-sparing effect in terms of future medication use, and GPi may provide superior control of dyskinesias as well as fewer alterations of mood, cognition, and behavior.


In addition to its reversibility, deep-brain stimulation appears safer than lesioning, even when bilateral. The treatment effect may be modulated; it causes less permanent collateral damage to the surrounding brain, without ruling out the possibility of new medical or surgical therapies as they become available. Drawbacks include increased time and expense, including specialized intraoperative and postoperative device management, the infection risk of indwelling hardware, potential magnetic resonance imaging (MRI) incompatibility, and the need for periodic battery replacement. Anesthesia for deep-brain stimulation is discussed later.


Future directions for surgical management include neural transplantation of dopaminergic cells, targeted infusion of glial cell line–derived neurotrophic factor (GDNF), gene therapy, and continuous duodenal levodopa infusion.


Anesthetic considerations


Preoperative Concerns


A thorough history of the patient’s symptoms, disease severity, and medication regimen should be obtained. The patient’s prescribed medication regimen should be optimized before scheduling surgery. Because several medications used to treat PD have a relatively short half-life, doses should be continued through the morning of surgery, and until as close to induction of anesthesia as feasible for afternoon or unscheduled procedures ( Box 8-5 ).



Box 8-5

Basal Ganglia and Cerebellar Disorders: Anesthetic Issues


Parkinson’s Disease (PD)





  • Medication effects are common and include motor fluctuations and dyskinesias, orthostatic hypotension, and antimuscarinic symptoms.



  • Deep-brain stimulation is a safe, reversible treatment for patients without cognitive impairment; implantation requires patient cooperation with intraoperative neurologic examination. Risks include air embolism and intracerebral hemorrhage.



  • Patients with PD undergoing any surgery should continue their medication regimen until immediately before surgery and should resume their medications as soon as possible postoperatively. Medications with potential extrapyramidal side effects should be avoided.



  • Patients with PD are particularly vulnerable to aspiration, respiratory compromise, and postoperative delirium.



Huntington’s Disease (HD)





  • Patients with advanced disease are at risk for aspiration and respiratory complications.



  • Patients with HD may have decreased pseudocholinesterase activity.



Sydenham’s Chorea (SC)





  • Preoperative evaluation must include ECG and evaluation for carditis and valvular disease.



  • Procedural sedation may be difficult because of patient movement and may be facilitated by the use of neuroleptics, benzodiazepines, or propofol.



  • Patients with SC should receive long-term prophylaxis against recurrent group A streptococcal pharyngitis.



Dystonias





  • Dystonic symptoms may complicate positioning and in severe cases, airway management.



  • Dystonic symptoms are often relieved by sedation.



  • Dystonic patients may be managed using anticholinergic or dopaminergic therapy, leading to possible drug-drug interactions.



  • There are no specific contraindications to any particular anesthetic agent, although many have been implicated in acute dystonic reactions.




Preoperative Evaluation


Cognitive impairment may predispose patients to postoperative delirium, and baseline mental status should be assessed. Neurologic symptoms, including tremor, muscle rigidity, and bradykinesia, should also be assessed. Patients with PD are particularly vulnerable to respiratory complications, and the preoperative evaluation should elicit evidence of swallowing dysfunction, retained or excessive oropharyngeal secretions, dyscoordination or rigidity of accessory muscles of respiration, and recent or active respiratory infection. From a cardiovascular standpoint, patients should be evaluated for arrhythmia and hypertension, as well as evidence of hypovolemia and orthostatic hypotension, which are common medication side effects. Autonomic dysfunction, including abnormal micturition, salivation, GI function, and temperature regulation should be considered. Seborrheic dermatitis of the face, ears, and skin folds is common in PD patients and should be recognized as a manifestation of underlying autonomic dysfunction. Weight loss and malnutrition are also common in patients with more advanced disease, and GI dysmotility and esophageal reflux disease can increase the risk of aspiration. Glucose utilization is often disrupted, and preoperative serum glucose should be measured.


Intraoperative Management


As suggested, intraoperative challenges in the management of patients with PD include increased aspiration risk, potential for autonomic instability and arrhythmia, and relatively high incidence of orthostatic hypotension. Tremor and rigidity may make positioning and cooperation difficult for patients undergoing sedation or preinduction vascular access or monitor placement. On the other hand, regional anesthesia may reduce the risk of aspiration and respiratory complications, minimize the risk of postoperative nausea and vomiting, and facilitate earlier ability to resume enteral medications. When tremor is a serious impediment to the safe performance of a procedure (e.g., ophthalmologic surgery), diphenhydramine may be a useful adjunct for reducing tremor in the awake or sedated patient.


In the absence of hypovolemia or cardiac dysfunction, propofol is a good choice for induction of general anesthesia because of its rapid metabolism and predictable offset. Although case reports have linked propofol to dyskinesias, it has also been noted to diminish tremor in the postoperative period. Thiopental has been associated with dyskinesias in several reports. Although ketamine may increase oral secretions and is relatively contraindicated because of its potential to cause hypertension through an exaggerated sympathetic response, it has been used without incident. Inhaled anesthetics have been associated with an increase in postoperative rigidity.


Nondepolarizing neuromuscular blockers may mask tremor, but patients with PD appear to have normal sensitivity to these drugs. Hyperkalemia has been reported in one patient with presumed denervation from chronic PD who received succinylcholine. However, a subsequent case series found no evidence of hyperkalemia in PD patients exposed to succinylcholine. Anticholinesterase agents have been used in the treatment of some parkinsonian symptoms and are presumed safe to use for the reversal of NMB. Because it does not cross the blood-brain barrier, glycopyrrolate is the preferred anticholinergic, particularly in patients with cognitive deficits.


Opioid medications should be used with caution in patients who are unlikely to tolerate respiratory depression. Fentanyl and remifentanil may exacerbate muscle rigidity, although this effect is amenable to treatment with neuromuscular blockers and may be blunted by coadministration of propofol. At low doses, morphine has been reported to decrease dyskinesias, but at high doses it may actually precipitate them. NSAIDs may be beneficial as part of an opioid-sparing strategy.


Medications that may precipitate extrapyramidal symptoms or dystonic reactions, including phenothiazines (promethazine), butyrophenones (droperidol), and metoclopramide are contraindicated in patients with PD. Benzodiazepines may diminish tremor, but may also exacerbate delirium and should be avoided in patients with significant dementia. Vasodilators should be used with caution in patients at risk for hypovolemia, and hypotension should be treated with volume resuscitation and direct-acting agents such as phenylephrine rather than ephedrine.


Postoperative Concerns


Patients with PD should be watched closely for postextubation respiratory failure. These patients are at increased risk of laryngospasm, inability to handle oral and respiratory secretions, and aspiration pneumonia. Respiratory reserve is likely to be diminished, especially in patients with preoperative muscle rigidity and dyscoordination. Intraoperative atelectasis and medication effects are likely to impair respiratory function further. Oral secretions may be a contraindication to noninvasive ventilation. In patients who fail to meet criteria for extubation or who are reintubated for respiratory failure, antiparksinonian medications should be continued via gastric tube to maximize chances of successful extubation.


Associated procedures


Anesthesia for Deep-Brain Stimulator Placement


Deep-brain stimulation (DBS) surgery is generally accomplished using two separate procedures. In the first stage, a stereotactic head frame is placed with the patient in a semi-upright position, and leads are then placed through burr holes using stereotactic guidance and patient cooperation. Several techniques have been used to facilitate lead placement; all share the goal of protecting the airway, providing adequate patient comfort, and allowing patient cooperation with intraoperative neurologic examination to optimize lead positioning. Monitored anesthesia care, scalp block, IV sedation, and general anesthesia using an asleep-awake-asleep technique have all been described. Maintenance of airway patency and ventilation are particularly important, given the difficulties of airway management once the head frame is in place. In addition to their potential for disinhibition or confusion in patients with cognitive deficits, the effects of GABAergic medications on electrophysiologic brain monitoring and their tendency to suppress tremor and rigidity may contraindicate their use.


Intraoperative risks include an increased potential for venous air embolism in the semi-upright position and intracerebral hemorrhage (2%-4% of DBS cases). Maintenance of systolic BP less than 140 mm Hg has been suggested in an effort to reduce the risk of hemorrhage. Stimulator implantation is usually performed 1 to 4 weeks after lead placement. Because neuromonitoring is not required, DBS is usually performed under general anesthesia and carries risks similar to permanent pacemaker implantation.


Anesthesia for Patients with Implantable Deep-Brain Stimulators


Anesthesia for patients with implantable deep-brain stimulators has not been well studied. MRI compatibility varies slightly by device, and the manufacturer should be consulted before proceeding with MRI in these patients. Electrocautery has the theoretic potential to cause generator malfunction and inappropriate electrode activation or CNS burns, but a recent review found no evidence of such complications. If possible, bipolar diathermy eliminates this concern. Scant evidence is available to guide intraoperative device management. Turning off the device intraoperatively seems a safe approach, but whether it is necessary is unclear. Postoperative akinesia may respond to levodopa, but some recommend turning the generator back “on” before emergence from anesthesia.


Huntington’s Disease (Chorea)


Huntington’s disease (HD; Huntington’s chorea) is an inherited progressive neurodegenerative condition whose hallmarks include choreoathetoid movements, dementia, and psychiatric disturbances. Although the exact mechanism has yet to be fully elucidated, HD appears to develop from the neurotoxicity of an abnormal variant of the protein huntingtin, arising from an autosomal dominant mutation involving a trinucleotide expansion (CAG) in the Huntington gene on chromosome 4p. Onset typically occurs in middle age, with the development of chorea, which is often accompanied by insidious progression of cognitive and personality changes. Choreatic movements are often subtle initially, but progress to the point of interference with normal movement. In late stages, chorea begins to affect the diaphragm and the muscles of the aerodigestive tracts, leading to dysphagia, dysarthria, and involuntary phonation. Onset and severity of the disease are influenced by the number of inherited trinucleotide repeats; because somatic instability can increase the number of repeats each generation, inheritance is subject to anticipation, the genetic phenomenon in which subsequent generations may experience earlier onset and worsened severity of the disease. Patients with juvenile-onset HD typically experience a rapidly progressive course. Prevalence of HD in Europe and North America has been estimated at 5 to 8 per 100,000 population.


Although the huntingtin protein is expressed throughout the body, it appears that only a subset of neurons are affected by the abnormal variant. The typical pathologic finding in patients with HD is diffuse atrophy of the caudate and putamen. Early onset is also associated with atrophy of cerebellar Purkinje cells. At the cellular level, cytoplasmic and intranuclear aggregates of the amino-terminus of mutant huntingtin are characteristic; whether these aggregates are causative or protective remains unclear. Huntingtin is necessary for normal embryonic development, and the presence of trinucleotide expansion does not seem to affect this process. In the adult, its role is less clear, although huntingtin is known to interact with multiple proteins and may play a role in protein trafficking, vesicular transport and anchoring, endocytosis, postsynaptic signaling, or cell survival. Conversely, the abnormal variant in HD may disrupt a wide variety of processes and pathways, including protein degradation, axonal transport, and synaptic transmission, and may lead to excitotoxicity, cellular metabolic dysfunction, and abnormal apoptosis.


To date, management of patients with HD remains supportive. Physical and occupational therapy, nutrition, and psychosocial counseling are important strategies for maximizing quality of life. Chorea may respond to treatment with tetrabenazine, which blocks dopamine transport. However, use of this agent may worsen bradykinesia, cognition, and mood. Neuroleptic agents may also be useful in controlling chorea as well as agitation and psychosis. Depression is common in patients with HD and may respond to tricyclic antidepressants (TCSs) or selective serotonin reuptake inhibitors (SSRIs). Recent studies have evaluated the possible disease-modifying effects of vitamin E, creatine, coenzyme Q10, highly unsaturated fatty acids (HUFAs), ethyl eicosapentaenoate (fatty acid derivative), minocycline (antiapoptotic), and riluzole (inhibitor of striatal glutamine). None of these agents has demonstrated a robust benefit, but trials are ongoing. Gene therapy, vaccination with the huntingtin protein, DBS, and surgical delivery of fetal tissue or neurotrophic factors have been proposed, and in some cases studied in animals or small human trials.


Anesthetic considerations


Data regarding anesthetic management of HD patients are limited. Patients with HD may have difficulty holding still during procedures performed with sedation (see Box 8-5 ). Butyrophenones and phenothiazines may alleviate choreiform movements, and benzodiazepines may be used to treat acute anxiety. However, caution should be used in patients with dementia, who are at increased risk for postoperative delirium. In advanced stages, patients with HD are vulnerable to dysphagia, aspiration, and respiratory complications. No clear contraindications seem to exist to the use of the common induction agents, neuromuscular blockers, or inhaled anesthetics. One case of delayed recovery from thiopental has been reported, but others have used this drug without incident. Patients with HD may have decreased pseudocholinesterase activity, but only a single case of delayed recovery from succinylcholine has been reported.


Associated procedures


Although there are no procedures specifically related to the treatment or management of HD, patients with advanced disease may require gastric feeding tube placement. These patients are also at high risk for falls. Orthopedic procedures, in particular hip and femur fixation, may be required.


Sydenham’s Chorea (Rheumatic Chorea)


Sydenham’s chorea (SC) is an acute manifestation of rheumatic fever, occurring in approximately one third of cases. Chorea is often asymmetric and occasionally unilateral, and it is generally accompanied by muscular weakness and emotional lability. Although the incidence of rheumatic fever has declined to 2 to 14 cases per 100,000 people in Europe and North America, almost 500,000 new cases occur each year, primarily in the nonindustrialized world. Children ages 5 to 13 years are most likely to be affected by SC, and girls are twice as likely to develop SC as boys. A familial predisposition to both acute rheumatic fever and to SC has been suggested. Onset of chorea is typically gradual, occurring 1 to 8 months after the inciting group A streptococcal infection. Emotional lability may precede motor symptoms, which often progress from subtle hand movements to irregular writhing movements of the arms. Ballistic movements, facial grimacing, tongue fasciculations, and diffuse hypotonia are also common. Psychiatric manifestations include irritability, inappropriate laughing or crying, and obsessive-compulsive behaviors. Symptoms, both motor and psychiatric, generally resolve in 3 to 4 months, and most patients recover completely. However, persistent symptoms have been described more than 2 years after onset, and up to 20% to 30% of SC patients have relapses, particularly those who do not receive antibiotic prophylaxis against recurrent streptococcal infection.


The pathogenesis of SC remains incompletely understood. Group A streptococcal infection stimulates the formation of antibodies against the N -acetyl-β- d -glucosamine (NABG or GlNAc) streptococcal antigen. These antibodies appear to cross-react with host antigens, leading to valvular injury in rheumatic endocarditis, for example. Some of these antibodies bind to lysoganglioside on the surface of neurons, initiating a calcium-mediated cell-signaling cascade. Others cross-react with tubulin, suggesting a link to the pathogenesis of other antibody-mediated motor neuropathies. At the gross anatomic level, small studies using MRI, positron emission tomography (PET), and single-photon emission computed tomography (SPECT) suggest that the basal ganglia and striatum are subject to reversible changes consistent with hypermetabolism and hyperperfusion.


Initial treatment of acute rheumatic fever involves antibiotic therapy to eliminate carriage of group A streptococci, anti-inflammatory therapy (aspirin) for patients with carditis, and heart failure management for those with severe valvular lesions. Evidence suggests that treatment with steroids may hasten resolution of SC. Numerous agents have been used in the pharmacologic management of chorea symptoms, including valproic acid, carbamazepine, haldol, benzodiazepines, and pimozide. Intravenous immune globulin (IVIG) and plasma exchange have also been used. All have shown benefit in small studies, but no conclusive evidence supports any of these treatments. Antibiotic prophylaxis against recurrent group A streptococcal pharyngitis is indicated for secondary prevention of recurrent episodes of rheumatic fever.


Anesthetic considerations


Preoperative evaluation of the patient with Sydenham’s chorea or a history of rheumatic fever should include electrocardiogram (ECG) and evaluation for endocarditis and valvular disease (see Box 8-5 ). As with other movement disorders, procedural sedation may be challenging, and symptomatic treatment of chorea with neuroleptics, benzodiazepines, or propofol may be helpful. A lack of published experience with SC patients precludes any list of contraindications other than those related to cardiac complications of acute rheumatic fever and possible interactions involving the drugs used for symptom management.


Dystonias


Dystonias are a broad group of movement disorders in which sustained involuntary muscle contractions lead to abnormal postures or repetitive movements. Classification schemes have centered on age of onset (early vs. late), anatomic distribution, and etiology (primary vs. secondary). Numerous genetic subtypes have also been identified. Prevalence estimates for dystonias as a group have varied widely. The most-often cited figures for the United States come from a 30-year study of Rochester, Minnesota, which found an incidence of 2 cases of generalized dystonia per 1 million persons per year, and 24 cases of focal dystonia per 1 million per year.


Etiology


Primary dystonias are generally characterized by a gradual onset and progression of symptoms, and occur in the absence of other neurologic, imaging, or laboratory abnormalities. Early-onset dystonias present in childhood or young adulthood, often beginning in one leg, and become generalized in a majority of patients. Late-onset dystonias (occurring in adulthood) typically present in the neck, arms, or face, and are likely to remain focal or segmental (involving contiguous body areas).


Cervical dystonia, also referred to as “spasmodic torticollis,” is the most common primary focal dystonia. Dystonia may involve rotation, lateral flexion, or anterior flexion of the head and is painful in 50% of patients. Dystonic contractions may lead to finer movements of the head that may resemble tremor. Oromandibular and facial dystonias involve the muscles of the jaw, tongue, and larynx and may cause difficulty with speech and swallowing. Spasmodic dysphonia refers to a focal dystonia of the laryngeal muscles, most often involving the adductor muscles, and therefore causing difficulty with vocalization rather than airway occlusion. Blepharospasm involves the periocular muscles and may impact vision when severe enough to cause prolonged involuntary eye closure. Blepharospasm may also occur in conjunction with oromandibular and facial dystonias, a constellation referred to as Brueghel’s or Meige’s syndrome. Focal or segmental dystonias of the upper extremity are also common. These are generally unilateral, are often absent at rest, and may produce an apparent tremor in addition to abnormal posturing. Repetitive performance of specific muscular tasks may produce occupational dystonias, such as writer’s cramp or the embouchure dystonia that has affected players of woodwind or brass instruments. An interesting feature of many focal dystonias is the presence of a geste antagoniste, or specific maneuver, such as a light touch to the overlying skin, which will relieve the dystonic contractions.


Several hereditary dystonias have been identified, and genetic predisposition or vulnerability likely plays a role in many cases. Mutation of the TOR1A gene, which encodes torsinA, an ATP-binding protein, may account for as many as half of all cases of early-onset primary dystonia. Other hereditary dystonias include dopa-responsive dystonia, an early-onset form often associated with parkinsonian features and notable for its responsiveness to levodopa therapy; various forms of dystonia plus parkinsonism; several types of paroxysmal dyskinesia; and myoclonus dystonia, an autosomal dominant condition involving myoclonic upper body jerks with dystonic features.


Secondary dystonias occur as a result of an alternative primary process. In these cases, dystonia is generally associated with additional neurologic, laboratory, or imaging abnormalities. Stroke, medication effects, and musculoskeletal or CNS trauma are common causes. Atypical presentations should also prompt further investigation. Numerous degenerative, genetic, and metabolic disease processes have been associated with dystonia; notable examples include Wilson’s, Huntington’s, and Leigh’s diseases; corticobasal degeneration; and cyanide or manganese toxicity.


Pathophysiology


Primary dystonias are not associated with specific neuropathology and do not appear to involve neuronal cell degeneration. They have also been difficult to localize anatomically. PET and functional MRI studies have suggested abnormal metabolic activity in the motor cortex and supplementary motor areas, the cerebellum, and the basal ganglia. DBS recordings have implicated abnormal function of the globus pallidus. Functionally, electrophysiologic studies suggest that dystonia involves loss of normal inhibitory signals, abnormal plasticity of the motor cortex, and subtle abnormalities of sensory function. The neurochemistry of dystonia remains poorly understood. The overlap between dystonia and parkinsonism, as well as the acute dystonic reactions that can accompany pharmacologic antagonism of dopamine receptors, suggest the importance of dopaminergic pathways in the neurochemistry of dystonia. The clinical observation that anticholinergic therapy can be effective in treating childhood dystonia suggests the importance of cholinergic pathways as well.


Treatment


Treatment of dystonia is symptomatic. Appropriately, dopa-responsive dystonia can be almost completely abolished with levodopa therapy. This response is generally sustained, and required doses are often small enough to prevent motor side-effects. Up to 15% of other dystonias will also respond to levodopa therapy. Anticholinergic drugs, including trihexyphenidyl and tetrabenazine, have also been widely used in the treatment of dystonia, but evidence for their efficacy is equivocal. Anecdotal evidence also suggests that some patients may benefit from benzodiazepines, baclofen, carbamazepine, zolpidem, or dopamine receptor blockers. Injection of affected muscle groups with botulinum toxin (BoNT-A and BoNT-B) has proved safe and effective and is now considered a first-line therapy for patients with cervical dystonia, blepharospasm, focal upper limb dystonias, and spasmodic dysphonia. Although sustained benefits are possible using botulinum toxin, not all patients will respond, and some will develop resistance to BoNT-A. These patients may respond to the alternate serotype BoNT-B, but some patients will fail treatment with either type. In patients with medication-resistant segmental or generalized primary dystonia, DBS of the globus pallidus has been effective in controlling symptoms for a majority of patients. Long-term follow-up data are limited. Although some evidence indicates a benefit in focal dystonia treated with DBS, data for the treatment of secondary dystonia are mixed.


Anesthetic considerations


Preoperative evaluation of the patient with dystonia should focus on the severity of the dystonic movements, known triggers or sensory tricks to break spasm, and potential interactions with levodopa or anticholinergic therapy (see Box 8-5 ). Although symptoms are often relieved with sedation, cervical or oropharyngeal dystonias may complicate airway management, and in severe cases, fiberoptic intubation techniques may be indicated for patients with limited mobility of the neck or jaw. Spasms are abolished by NMB and also appear to be relieved by inhaled nitrous oxide (N 2 O) concentrations greater than 50%. No anesthetic agents are contraindicated, although there are numerous case reports of acute dystonia during and immediately after general anesthesia using various techniques including N 2 O/sevoflurane, N 2 O/propofol, and propofol/fentanyl/vecuronium.




Diseases of myelin


The myelin diseases are a large group of neurodegenerative disorders associated with abnormal myelinization of either the central or the peripheral nervous system and also referred to as “demyelinating diseases.” This group can be subdivided into dysmyelinating and demyelinating disorders. The dysmyelinating subgroup includes disorders associated with inherited defective production of myelin, often manifesting at birth, whereas demyelinating disorders are acquired later in life and are characterized by a loss of normal myelin ( Box 8-6 ). Regardless of the exact cause, abnormal myelinization of the nervous system leads to impairment of nerve conduction and eventual loss of function in the affected nerves across the whole spectrum of the nervous system. Although different disorders in this group may vary significantly in their presentation, all tend to have a broad range of symptoms involving many sensory, motor, and cognitive functions, which usually progress with time, leading to various degrees of disability.



Box 8-6

Diseases of Myelin


Central Nervous System





  • Multiple sclerosis



  • Leukodystrophies



  • Central pontine myelinosis



  • Subacute combined degeneration



  • Tabes dorsalis



  • Multifocal leukoencephalitis



  • Devic’s disease



  • Acute disseminated encephalomyelitis



Peripheral Nervous System





  • Hereditary primary motor sensory neuropathies



  • Acute (Guillain-Barré syndrome) or chronic inflammatory demyelinating polyneuropathy



Demyelinating (Acquired)





  • Multiple sclerosis



  • Central pontine myelinosis



  • Subacute combined degeneration (vitamin B 12 deficiency)



  • Tabes dorsalis



  • Acute disseminated encephalomyelitis



  • Progressive multifocal leukoencephalopathy



  • Mercury intoxication



Dysmyelinating (Congenital/Hereditary)





  • Hereditary primary motor sensory neuropathies



  • Leukodystrophies




    • Krabbe’s disease



    • Alexander’s disease



    • Pelizaeus-Merzbacher disease



    • Canavan’s disease



    • Others





For most of the myelin disorders, no anesthesia experiences have been reported. Therefore the focus here is on the most common conditions in this group for which a sufficient number of reports address various aspects of perioperative care in these patients. The same anesthetic considerations could be extrapolated when providing anesthesia care for patients with similar conditions.


Multiple Sclerosis


Multiple sclerosis (MS) is an acquired inflammatory autoimmune disorder possibly caused by an interplay of genetic and environmental factors, although its exact etiology is unknown. MS is characterized by widespread, initially partially reversible, demyelination of axonal sheaths in the CNS and subsequent development of sclerotic lesions in the brain, which eventually results in permanent neurodegeneration. MS presents with a wide variety of signs and symptoms involving sensory, motor, autonomic, and cognitive functions ( Box 8-7 ). MS characteristically first manifests in patients in their 20s to 40s, with a twofold to threefold higher prevalence in women. Many of these patients develop severe disability over time, and many die from complications related to MS. The etiology, epidemiology, pathophysiology, clinical presentation, and treatment of MS have been recently reviewed.



Box 8-7

Multiple Sclerosis: Clinical Signs/Symptoms *

* From most common to less common.



Sensory Symptoms





  • Numbness, “pins and needles,” tingling in limbs



  • Swelling, tightness, coldness of limbs



  • Proprioceptive deficits



  • Facial sensory symptoms



Ophthalmic Symptoms





  • Visual loss related to optic neuritis



  • Diplopia



  • Internuclear ophthalmoplegia



Motor Symptoms





  • Subacute paraparesis or paraplegia, more in lower extremities



  • Spasticity and possible contractures



Coordination





  • Vertigo



  • Gait imbalance



  • Limb ataxia



Autonomic Nervous System Dysfunction





  • Bladder, bowel, and sexual dysfunction



  • Possible orthostatic hypotension



Other Symptoms/Signs





  • Temperature and exercise insensitivity (Uhthoff phenomenon or heating reaction)



  • Pain



  • Fatigue



  • Depression



  • Seizures




None of the signs or symptoms listed in Box 8-7 is diagnostic of or unique to MS, which can present with neurologic symptoma found in other nervous system disorders. The combination of these symptoms and the clinical course of MS progression eventually leads to the diagnosis. The following subtypes of the disease progression have been identified:




  • The relapsing-remitting subtype is characterized by acute onset of new symptoms (relapses, or exacerbations) followed by long periods of remission, during which most symptoms tend to abate or completely reverse. Timing of relapses is unpredictable, and often no precipitating factor can be identified. Up to 80% of patients have this pattern as initial presentation of MS.



  • Secondary progressive MS is characterized by progression of the clinical picture without periods of obvious remission. Many patients with relapsing-remitting disease eventually convert to secondary progressive MS.



  • The primary progressive subtype is found in patients whose clinical symptomatology continues to accumulate without significant improvements in symptoms after initial onset of MS.



  • The progressive-relapsing subtype is the rarest form and is characterized by steady, progressive development of disability with superimposed acute episodes of new neurologic symptoms.



The MS progression patterns may have a significant effect on the decision making in the perioperative management of these patients. Although relapses in patients with near-complete remission or sudden worsening of existing symptoms can be precipitated by perioperative events, they are not necessarily synonymous with poor prognosis, but have a powerful emotional effect on MS patients and may dramatically, even if only temporarily, affect their quality of life.


Factors clinically established as exacerbating MS include stressful events such as emotional and physical trauma, infections, surgery, and the peripartum period. Hormonal and temperature fluctuations appear to be clinically correlated with exacerbations as well. Even small elevations in body core temperature above normal, as occur in the perioperative setting, can block nerve conduction in previously demyelinated nerve fibers and produce new neurologic deficits or worsen others. Previously, Edmund and Fog documented clinical deterioration with temperature elevations in 75% of MS patients. However, it is important to emphasize that no evidence indicates that exposure to these precipitating factors affects the overall prognosis of the disease in these patients.


Treatment


Current therapies for MS are not curative but attempt to ameliorate acute or chronic symptoms and disability and reduce the number of future exacerbations. Most MS relapses respond to glucocorticoid therapy in the early phases, but many patients fail to respond to corticosteroids as their number of exacerbations increases. Patients with optic neuritis respond to oral and IV adrenocorticotropic hormone (ACTH) and prednisone in the acute phases and experience fewer relapses. Other immunosuppressive agents, such as cyclophosphamide, cytarabine, and azathioprine, have shown intermittent success at preventing the number and severity of relapses, but do carry side effects. Patients with severe motor spasticity and bladder dysfunction are treated with antispastic medications, which provide some relief. Alternative therapies not clinically proven but tried in severely resistant cases include plasmapheresis, hyperbaric oxygen therapy, linoleate supplementation, and interferons. The most recent, controversial development is related to the “vascular hypothesis,” suggesting that MS could be caused by the chronic cerebrospinal venous insufficiency frequently found in these patients. Cerebral venous balloon angioplasty result in significant improvement in the course of MS in preliminary reports. The results of these series are inconclusive until proper blinded randomized studies can be conducted. However, if this treatment were to gain a wider acceptance, administering anesthesia for this procedure may significantly increase exposure to patients with MS among anesthesiologists.


Anesthetic considerations


Case reports and observational studies are limited regarding anesthesia effects and perioperative management in patients with multiple sclerosis. Some of the earlier reports implicated a possible connection between the use of general or neuroaxial anesthesia and relapse of MS or worsening of existing symptoms. Despite these reports providing scant conclusive evidence to support this claim, it has been difficult to disprove this connection because of the unpredictable nature of relapses and the potential for the temporary worsening of existing neurologic deficits, even in the absence of a new demyelinating event. This worsening can be caused by relatively benign perioperative derangements of homeostasis, such as hyperthermia or hyponatremia, or administration of anesthetic agents. This usually brief deterioration is most likely caused by an enhanced effect of these conditions on already-impaired but clinically silent conduction delay in previously demyelinated nerves. Current opinion in anesthesia literature does not support the idea that administration of anesthesia can be linked to a new demyelinating event in patients with MS. In the absence of large, controlled prospective studies, it is impossible to resolve this issue. Given this lack of conclusive evidence, designing an anesthetic plan for patients with MS, even currently asymptomatic, poses a number of unique challenges ( Box 8-8 ).



Box 8-8

Multiple Sclerosis (MS): Anesthetic Issues





  • Patients with MS have an unpredictable pattern of relapse that may coincide with postoperative recovery.



  • Informed consent regarding the effect of anesthetic agents on the course of the disease is difficult to provide due to lack of conclusive evidence in the literature.



  • Thorough preoperative interview and maximal patient participation regarding choice of anesthetic technique is advisable, combined with detailed documentation of these discussions.



  • Neuroaxial and regional anesthesia are not contraindicated in MS patients, but lower concentrations of local anesthetic and careful titrations are advisable.



  • Epidural anesthesia may be preferable to spinal anesthesia.



  • Patients with MS are at increased risk of intraoperative autonomic dysfunction.



  • Patients with advanced disease are at increased risk of perioperative respiratory depression and sleep apnea.



  • Avoid monitoring neuromuscular blockade at the affected muscles to avoid overdose.



  • Use of depolarizing muscle relaxants should be avoided when possible in MS patients.




Patients with MS presenting for surgery and anesthesia may typically fit a number of distinct clinical scenarios, which require different approaches to perioperative management. In patients with the relapsing-remitting pattern in a remission presenting for elective or emergency surgery, the main concern is a possible postoperative relapse, either precipitated by perioperative events or merely coincidental. It is of particular importance to inform these often asymptomatic but fearful patients regarding the lack of conclusive evidence connecting the use of anesthetic agents and MS exacerbation episodes.


Thorough preoperative interview, detailed anesthesia consent, and maximal engagement in the decision-making process regarding the choice of anesthetic technique and agents are crucial in this group of patients. Asymptomatic pregnant patients with a history of MS pose a special challenge regarding the use of epidural or spinal anesthesia for labor or cesarean section. Because MS mainly affects women in their childbearing years, many of these women will likely need anesthesia for cesarean birth or ask for analgesia for labor. A further complication is that late pregnancy is associated with relief of MS symptoms and reduced number of relapses. However, the possibility of postpartum relapse increases compared with nonpregnant patients, although pregnancy does not seem to adversely affect the overall prognosis of the disease.


Use of neuraxial blockade in patients with MS is controversial because of the potential for direct neurotoxicity exerted by local anesthetics, especially on demyelinated nerves. Local and regional anesthesia remains a controversial topic in MS patients because of the potential pharmacodynamic effects on nerve conduction. With the pathology of demyelination in MS, one might expect that the potential for neurotoxicity with local anesthetics administered in the epidural or intrathecal space would be higher. However, several retrospective studies have not demonstrated a significant increase in MS exacerbations with either epidural or spinal local anesthetic administration. Although unproved, many clinicians believe that repeated doses of local anesthetics for regional anesthesia carry a potentially increased risk for neurotoxicity, both locally and centrally, because the blood-brain barrier may be more permeable as a result of chronic inflammatory changes. Therefore, many believe that lower concentrations of local anesthetics should be used and combined with narcotics, because there have been no reports of significant adverse events with epidural or intrathecal opioids. In a few anecdotal reports, regional or neuroaxial anesthesia precipitated MS episodes in previously undiagnosed patients, and temporary worsening of symptoms followed neuroaxial block for labor.


The prevailing view is that administration of spinal anesthetic exposes the spinal cord to higher, potentially toxic concentrations compared with epidural anesthesia. Despite these concerns and the limited evidence available, the existing literature largely supports the view that use of regional anesthesia does not lead to true relapses of MS and should be strongly considered in willing patients. This topic has been recently reviewed.


The paucity of conclusive evidence regarding the effects of local and regional anesthesia on the course of MS, combined with a higher probability of postpartum relapse requires very thorough prenatal discussion of anesthetic choice, including careful documentation of these discussions. A recent survey of practicing anesthesiologists in the United Kingdom indicates a majority would perform epidural or spinal anesthesia in pregnant MS patients, provided special consideration were given to the relevant issues.


In a patient with the relapsing-remitting pattern scheduled for an elective procedure who is found to have new neurologic deficits, it is prudent to delay surgery for neurologic consultation and therapeutic intervention. In patients with progressive patterns of MS, anesthetic considerations are generally governed by the disease progression and severity, associated degree of disability, and comorbidities. Patients with advanced disease should be evaluated for signs of autonomic dysfunction, which would warrant the use of invasive monitoring to guide hemodynamic support intraoperatively. Compensatory mechanisms in response to intraoperative events, anesthetic agents, and vasopressors may be altered in these patients. Patients with cranial nerve involvement and history of upper airway obstruction and compromised respiratory function should be considered for postoperative ventilatory support in the ICU. Intraoperative positioning of patients with spasticity and contractures can be challenging and may require advance planning. Increases in body temperature are known to lead to dramatic increases in symptoms and may precipitate an onset of new symptoms. As a result, hyperthermia must be carefully avoided. Even mild fevers should be actively pre-empted and aggressively treated when detected.


Anesthetic induction agents and inhaled gases have no demonstrable adverse effects on nerve conduction and have not been implicated in the literature as contributing to MS progression. Drugs often administered under anesthesia, including anticholinergics, atropine, and glycopyrrolate, may also lead to increases in body temperature and should be used with caution. The use of depolarizing muscle relaxants also carries theoretic risks in MS patients, more specifically those with profound neurologic deficits that often cause upregulation of motor-end-plate acetylcholine receptors and the hyperkalemic response to depolarization. We have not found any studies that implicate nondepolarizing muscle relaxants in neurologic sequelae perioperatively; thus their use appears to be safe. Drugs used to treat MS and associated disorders need to be taken into consideration, especially corticosteroids and antiepileptic medications.


Nitrous Oxide–Induced Subacute Combined Degeneration


Subacute combined degeneration (SCD) is a neurodegenerative demyelinating disorder of the spinal cord observed in patients with vitamin B 12 deficiency usually suffering from pernicious anemia, various conditions leading to malnutrition, tropical sprue, or HIV infection. A wide range of neurologic symptoms consistent with myelopathy, such as progressive weakness, sensory ataxia, paresthesias, spasticity, paraplegia, incontinence, dementia, and visual loss result from progressive degeneration of the dorsal and lateral white matter of the spinal cord and in the brain. Neurologic symptoms may occur in patients without megaloblastic anemia. This condition is of particular interest to an anesthesiologist because of the unique role that N 2 O can play in the development of this condition. By oxidizing the cobalt in the cobolamin, N 2 O inactivates vitamin B 12 and consequently reduces methionine synthase activity. Inadequate production of methionine leads to failure of myelin maintenance in spinal cord and axonal degeneration.


Subacute combined degeneration has been reported in previously healthy recreational N 2 O abusers with normal blood levels of B 12 . However, even single use of N 2 O for anesthesia in asymptomatic patients with subclinical deficiency of vitamin B 12 may lead to delayed SCD development. Repetitive and prolonged exposure to N 2 O in patients with B 12 deficiency increases the probability of SCD development. Additionally, SCD tends to develop days or even weeks after uncomplicated anesthesia with N 2 O, rendering it difficult to make a connection between the use of N 2 O and the progressive neurologic deterioration. Classic signs of developing myelopathy, often in combination with personality changes and intellectual impairments, after recent surgery in patients with pre-existing disorders predisposing to vitamin B 12 deficiency should assist in arriving at correct diagnosis.


Prompt treatment with vitamin B 12 and methionine supplementation usually lead to rapid stabilization and often gradual improvement and although not necessarily complete, reversal of symptoms. In patients with known vitamin B 12 deficiency or predisposing conditions such as malnutrition, gastritis, or gastrectomy, N 2 O should not be used for anesthesia. With the availability of newer inhalational and intravenous agents characterized by low context-sensitive decrement times, N 2 O can be easily replaced without sacrificing the speed of emergence from general anesthesia.




Peripheral nerve disease and the polyneuropathies


Peripheral nerve disease covers a wide variety of causation and clinical entities ( Box 8-9 ). The peripheral nervous system (PNS), which encompasses all neural structures outside the spinal cord and brainstem, includes a broad variety of cell types, nerve fibers, anatomic variability, and function. Likewise, the pathologic conditions capable of affecting the PNS at multiple points also vary greatly. Signs and symptoms of polyneuropathies can therefore include impaired motor function, spasm and fasciculations, reflex changes, sensory loss, pain and paresthesias, dysesthesias, ataxia, tremor, trophic changes, and autonomic dysfunction. These can all have acute or chronic onset and duration.


Sep 5, 2019 | Posted by in ANESTHESIA | Comments Off on Neurologic Diseases

Full access? Get Clinical Tree

Get Clinical Tree app for offline access