Sei sulla pagina 1di 22

10/26/2018 https://emedicine.medscape.

com/article/315632-print

emedicine.medscape.com

Guillain-Barre Syndrome
Updated: May 04, 2018
Author: Michael T Andary, MD, MS; Chief Editor: Milton J Klein, DO, MBA

Overview

Practice Essentials
Guillain-Barré syndrome (GBS) can be described as a collection of clinical syndromes that manifests as an acute inflammatory polyradiculoneuropathy with resultant weakness and diminished
reflexes.

Although the classic description of GBS is that of a demyelinating neuropathy with ascending weakness, many clinical variants have been well documented in the medical literature.

Signs and symptoms

The typical patient with GBS, which in most cases will manifest as acute inflammatory demyelinating polyradiculoneuropathy (AIDP), presents 2-4 weeks following a relatively benign respiratory
or gastrointestinal illness with complaints of finger dysesthesias and proximal muscle weakness of the lower extremities. The weakness may progress over hours to days to involve the arms,
truncal muscles, cranial nerves, and muscles of respiration.

Common complaints associated with cranial nerve involvement in GBS include the following:

Facial droop (may mimic Bell palsy)

Diplopias

Dysarthria

Dysphagia

Ophthalmoplegia

Pupillary disturbances

Most patients complain of paresthesias, numbness, or similar sensory changes. Paresthesias generally begin in the toes and fingertips, progressing upward but generally not extending beyond
the wrists or ankles.

Pain associated with GBS is most severe in the shoulder girdle, back, buttocks, and thighs and may occur with even the slightest movements. The pain is often described as aching or throbbing
in nature.

Autonomic changes in GBS can include the following:

Tachycardia

Bradycardia

Facial flushing

Paroxysmal hypertension

Orthostatic hypotension

Anhidrosis and/or diaphoresis

Urinary retention

Typical respiratory complaints in GBS include the following:

Dyspnea on exertion

Shortness of breath

Difficulty swallowing

Slurred speech

Ventilatory failure with required respiratory support occurs in up to one third of patients at some time during the course of their disease.

See Clinical Presentation for more detail.

Diagnosis

GBS is generally diagnosed on clinical grounds. A basic peripheral neuropathy workup is recommended in cases in which the diagnosis is uncertain. Biochemical screening can also be
conducted and would include the following studies:

Electrolyte levels

Liver function tests (LFTs)

Creatine phosphokinase (CPK) level

Erythrocyte sedimentation rate (ESR)

Needle EMG and nerve conduction studies

Signs of demyelination can include the following:

Nerve conduction slowing

Prolongation of the distal latencies

https://emedicine.medscape.com/article/315632-print 1/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
Prolongation of the F-waves[1, 2]

Conduction block or dispersion of responses: Evidence frequently demonstrated at sites of natural nerve compression

Weak muscles showing reduced recruitment: Demonstrated with needle examination (electromyography [EMG])

Pulmonary function

Maximal inspiratory pressures and vital capacities are measurements of neuromuscular respiratory function and predict diaphragmatic strength. Maximal expiratory pressures also reflect
abdominal muscle strength. Negative inspiratory force (NIF) is a relatively easy bedside test to measure respiratory muscle function. Normal is usually greater than 60 cm water. If the NIF is
dropping or nears 20 cm water, respiratory support needs to be available.

Cerebrospinal fluid studies

Most, but not all, patients with GBS have an elevated cerebrospinal fluid (CSF) protein level (>400 mg/L), with normal CSF cell counts. Elevated or rising protein levels on serial lumbar punctures
and 10 or fewer mononuclear cells/mm3 strongly support the diagnosis.

See Workup for more detail.

Management

Intensive care unit

Admission to the intensive care unit (ICU) should be considered for all patients with labile dysautonomia, a forced vital capacity of less than 20 mL/kg, or severe bulbar palsy.[3, 4] Any patients
exhibiting clinical signs of respiratory compromise to any degree also should be admitted to an ICU.[3]

Competent intensive care includes the following features:

Respiratory therapy

Cardiac monitoring

Safe nutritional supplementation

Monitoring for infectious complications (eg, pneumonia, urinary tract infections, septicemia)

Subcutaneous unfractionated or low ̶ molecular-weight heparin (LMWH) and thromboguards are often used in the treatment of immobile patients to prevent lower-extremity deep venous
thrombosis (DVT) and consequent pulmonary embolism (PE).

Immunomodulation

Immunomodulatory treatment in GBS has been used to hasten recovery. Intravenous immunoglobulin (IVIG) and plasma exchange have proved equally effective.

Physical, occupational, and speech therapy

Addressing upright tolerance and endurance may be a significant issue during the early part of physical rehabilitation. Active muscle strengthening can then be slowly introduced and may include
isometric, isotonic, isokinetic, or progressive resistive exercises.

Occupational therapy professionals should be involved early in the rehabilitation program to promote positioning, posture, upper body strengthening, range of motion (ROM), and activities that
aid functional self care.

Speech therapy is aimed at promoting speech and safe swallowing skills for patients who have significant oropharyngeal weakness with resultant dysphagia and dysarthria.

See Treatment and Medication for more detail.

Background
Guillain-Barré syndrome (GBS) can be described as a collection of clinical syndromes that manifests as an acute inflammatory polyradiculoneuropathy with resultant weakness and diminished
reflexes. With poliomyelitis under control in developed countries, GBS is now the most important cause of acute flaccid paralysis. (See Clinical Presentation.)

Although the classic description of GBS is that of a demyelinating neuropathy with ascending weakness, many clinical variants have been well documented in the medical literature, and variants
involving the cranial nerves or pure motor involvement and axonal injury are not uncommon. (See Pathophysiology.)

Acute inflammatory demyelinating polyradiculoneuropathy (AIDP) is the most widely recognized form of GBS in Western countries, but the variants known as acute motor axonal neuropathy
(AMAN), acute motor-sensory axonal neuropathy (AMSAN), and Miller-Fisher syndrome also are well recognized.

Based on a clinical spectrum of symptoms and findings, it is widely believed that strictly defined subgroups of GBS exist. However, these subgroups are not easily distinguished.

GBS remains a diagnosis made primarily through the assessment of clinical history and findings (see Clinical Presentation). Serum autoantibodies are not measured routinely in the workup of
GBS, but results may be helpful in patients with a questionable diagnosis or a variant of GBS (see Workup).

Approximately one third of patients require admission to an intensive care unit (ICU), primarily because of respiratory failure. After medical stabilization, patients can be treated on a general
medical/neurologic floor, but continued vigilance remains important in preventing respiratory, cardiovascular, and other medical complications. Treatment with intravenous immunoglobulin (IVIG)
or plasma exchange may hasten recovery. After discharge, outpatient physical therapy and occupational therapy may be beneficial in helping patients with GBS to regain their baseline functional
status. (See Treatment and Medication.)[5, 3, 6, 7]

Historical background

In 1859, Landry published a report on 10 patients with an ascending paralysis.[8] Subsequently, in 1916, 3 French physicians (Guillain, Barré, and Strohl) described 2 French soldiers with motor
weakness, areflexia, cerebrospinal fluid (CSF) albuminocytologic dissociation, and diminished deep tendon reflexes.[1] The identified syndrome was later named Guillain-Barré syndrome.
Historically, GBS was a single disorder; however, current practice acknowledges several variant forms.

Pathophysiology
GBS is a postinfectious, immune-mediated disease. Cellular and humoral immune mechanisms probably play a role in its development. Most patients report an infectious illness in the weeks
prior to the onset of GBS. Many of the identified infectious agents are thought to induce production of antibodies that cross-react with specific gangliosides and glycolipids, such as GM1 and
GD1b, that are distributed throughout the myelin in the peripheral nervous system.[9]

The pathophysiologic mechanism of an antecedent illness and of GBS can be typified by Campylobacter jejuni infections.[10, 11] The virulence of C jejuni is thought to be based on the presence
of specific antigens in its capsule that are shared with nerves.

Immune responses directed against lipopolysaccharide antigens in the capsule of C jejuni result in antibodies that cross-react with ganglioside GM1 in myelin, resulting in immunologic damage to
the peripheral nervous system. This process has been termed molecular mimicry.[12, 13]

Pathologic findings in GBS include lymphocytic infiltration of spinal roots and peripheral nerves (cranial nerves may be involved as well), followed by macrophage-mediated, multifocal stripping of
myelin. This phenomenon results in defects in the propagation of electrical nerve impulses, with eventual absence or profound delay in conduction, causing flaccid paralysis. Recovery is typically
associated with remyelination.

https://emedicine.medscape.com/article/315632-print 2/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
In some patients with severe disease, a secondary consequence of the severe inflammation is axonal disruption and loss. A subgroup of patients may have a primary immune attack directly
against nerve axons, with sparing of myelin. The clinical presentation in these patients is similar to that of the principal type.

Subtypes of Guillain-Barré syndrome

Several variants of GBS are recognized. These disorders share similar patterns of evolution, symptom overlap, and probable immune-mediated pathogenesis. Recovery from them varies.

Acute inflammatory demyelinating polyneuropathy

The acute inflammatory demyelinating polyneuropathy (AIDP) subtype is the most commonly identified form in the United States. It is generally preceded by a bacterial or viral infection. Nearly
40% of patients with AIDP are seropositive for C jejuni. Lymphocytic infiltration and macrophage-mediated peripheral nerve demyelination is present. Symptoms generally resolve with
remyelination.

Acute motor axonal neuropathy

The acute motor axonal neuropathy (AMAN) subtype is a purely motor disorder that is more prevalent in pediatric age groups.[14] AMAN is generally characterized by rapidly progressive
symmetrical weakness and ensuing respiratory failure.

Nearly 70-75% of patients with AMAN are seropositive for Campylobacter, with the majority of cases of AMAN being associated with preceding C jejuni diarrhea. Patients typically have high titers
of antibodies to gangliosides (ie, GM1, GD1a, GD1b). Inflammation of the spinal anterior roots may lead to disruption of the blood-CNS barrier.[12] Biopsies show wallerianlike degeneration
without significant lymphocytic inflammation.

Many cases have been reported in rural areas of China, especially in children and young adults during the summer months.[15] Pure axonal cases may occur more frequently outside of Europe
and North America. AMAN cases may also be different from cases of axonal GBS described in the West.

Prognosis is often quite favorable. Although recovery for many is rapid, severely disabled patients with AMAN may show improvement over a period of years.[16]

One third of patients with AMAN may actually be hyperreflexic. Although the mechanism for this hyperreflexia is unclear, dysfunction of the inhibitory system via spinal interneurons may increase
motor neuron excitability. Hyperreflexia is significantly associated with the presence of anti-GM1 antibodies.[8]

Acute motor-sensory axonal neuropathy

Acute motor-sensory axonal neuropathy (AMSAN) is a severe acute illness differing from AMAN in that it also affects sensory nerves and roots.[17] Patients are typically adults. AMSAN often
presents as rapid and severe motor and sensory dysfunction. Marked muscle wasting is characteristic, and recovery is poorer than it is from electrophysiologically similar cases of AMAN.

As with AMAN, AMSAN is often associated with preceding C jejuni diarrhea. Pathologic findings show severe axonal degeneration of motor and sensory nerve fibers with little demyelination.[18]

Miller-Fisher syndrome

Miller-Fisher syndrome (MFS), which is observed in about 5% of all cases of GBS, classically presents as a triad of ataxia, areflexia, and ophthalmoplegia.[19] Acute onset of external
ophthalmoplegia is a cardinal feature.[12] Ataxia tends to be out of proportion to the degree of sensory loss. Patients may also have mild limb weakness, ptosis, facial palsy, or bulbar palsy.
Patients have reduced or absent sensory nerve action potentials and absent tibial H reflex.[20]

Anti-GQ1b antibodies are prominent in MFS, and have a relatively high specificity and sensitivity for the disease.[21] Dense concentrations of GQ1b ganglioside are found in the oculomotor,
trochlear, and abducens nerves, which may explain the relationship between anti-GQ1b antibodies and ophthalmoplegia. Patients with acute oropharyngeal palsy carry anti-GQ1b/GT1a IgG
antibodies.[12] Recovery generally occurs within 1-3 months.

Acute panautonomic neuropathy

Acute panautonomic neuropathy, the rarest GBS variant, involves the sympathetic and parasympathetic nervous systems. Patients have severe postural hypotension, bowel and bladder
retention, anhidrosis, decreased salivation and lacrimation, and pupillary abnormalities. Cardiovascular involvement is common, and dysrhythmias are a significant source of mortality. Significant
motor or sensory involvement is lacking. Recovery is gradual and often incomplete.

Pure sensory GBS

A pure sensory variant of GBS has been described in the literature. It is typified by a rapid onset of sensory loss, sensory ataxia, and areflexia in a symmetrical and widespread pattern. Lumbar
puncture studies show albuminocytologic dissociation in the CSF, and results from electromyography (EMG) show characteristic signs of a demyelinating process in the peripheral nerves.

The prognosis in pure GBS is generally good. Immunotherapies, such as plasma exchange and the administration of IVIGs, can be tried in patients with severe disease or slow recovery.

Other variants

The pharyngeal-cervical-brachial variant of GBS is distinguished by isolated facial, oropharyngeal, cervical, and upper limb weakness without lower limb involvement. There can be combinations
of any of the above subtypes, and virtually any combination of nerve injury. There are likely mild cases that cause temporary symptoms, improve spontaneously, and never get definitively
diagnosed.

Other unusual clinical variants with restricted patterns of weakness are observed only in rare cases.

Etiology
GBS is considered to be a postinfectious, immune-mediated disease targeting peripheral nerves. Up to two thirds of patients report an antecedent bacterial or viral illness prior to the onset of
neurologic symptoms.[22, 23] Respiratory infections are most frequently reported, followed by gastrointestinal infections.[24] Administration of certain vaccinations and other systemic illnesses
have also been associated with GBS. Case reports exist regarding numerous medications and procedures; however, whether any causal link exists is unclear.

C jejuni

In several studies, C jejuni was the most commonly isolated pathogen in GBS. Serology studies in a Dutch GBS trial identified 32% of patients as having had a recent C jejuni infection, while
studies in northern China documented infection rates as high as 60%.[15, 25, 26]

Gastrointestinal and upper respiratory tract symptoms can be observed with C jejuni infections. C jejuni infections can also have a subclinical course, resulting in patients with no reported
infectious symptoms prior to the development of GBS. Patients who develop GBS following an antecedent C jejuni infection often have a more severe course, with rapid progression and a
prolonged, incomplete recovery. A strong clinical association has been noted between C jejuni infections and the pure motor and axonal forms of GBS.

The virulence of C jejuni is thought to result from the presence of specific antigens in its capsule that are shared with nerves. Immune responses directed against capsular lipopolysaccharides
produce antibodies that cross-react with myelin to cause demyelination.

C jejuni infections also generate anti-ganglioside antibodies—including to the gangliosides GM1, GD1a, GalNac-GD1a, and GD1b—that are commonly found in patients with AMAN and AMSAN,
the axonal subtypes of GBS. (Patients with C. jejuni enteritis not complicated by GBS, however, do not produce the specific anti-ganglioside antibodies.)

Even in the subgroup of patients with GM1 antibodies, however, the clinical manifestations vary. Host susceptibility is probably one determinant in the development of GBS after infectious illness.
[25, 27]

Although GM1 antibodies can also be found in patients with demyelinating GBS, they are much less common in these cases. C. jejuni infection can also generate antibodies to the ganglioside
GQ1b, a component of oculomotor nerve myelin; these are associated with MFS.

CMV

Cytomegalovirus (CMV) infections are the second most commonly reported infections preceding GBS, with CMV being the most common viral trigger of GBS. The aforementioned Dutch GBS
study found CMV to be present in 13% of patients.[28]

https://emedicine.medscape.com/article/315632-print 3/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
CMV infections present as upper respiratory tract infections, pneumonias, and nonspecific, flulike illnesses. GBS patients with preceding CMV infections often have prominent involvement of the
sensory and cranial nerves. CMV infections are significantly associated with antibodies against the ganglioside GM2.

Other infections

Other significant, although less frequently identified, infectious agents in GBS patients include Epstein-Barr virus (EBV), Mycoplasma pneumoniae, and varicella-zoster virus.[29] An association
between GBS and acute human immunodeficiency virus (HIV) infection also is well recognized.[8, 30, 31, 32, 33, 34]

Infections with Haemophilus influenzae, Borrelia burgdorferi, para-influenza virus type 1, influenza A virus, influenza B virus, adenovirus, and herpes simplex virus have been demonstrated in
patients with GBS, although not more frequently than they have in controls.[35]

There has been speculation that the Zika virus can cause GBS. Reported cases of the syndrome began to increase in Brazil during the Zika virus outbreak that was identified there in 2015, with
hundreds of cases of GBS reported that year.[36, 37] In July 2015, for example, out of 76 patients in the state of Bahia identified with neurologic syndromes, 42 were confirmed as having GBS,
with the symptom history of 26 of these confirmed cases having been consistent with Zika virus infection.[38]

Other Latin American countries to which the Zika outbreak spread, including Colombia, Venezuela, and El Salvador, also reported increases in GBS cases.[37] In El Salvador, where an average
of 14 cases of GBS are reported per month, 46 cases were reported between December 1, 2015 and January 6, 2016.[38] A literature review by Barbi et al estimated that 1.23% of Zika virus
cases could develop into GBS.[39]

However, additional research will be required to resolve speculation about the Zika-GBS link. The reliability of Zika virus diagnoses outside of the United States is not known. In the United States,
the only infectious disease laboratories capable of making this diagnosis are at the US Centers for Disease Control and Prevention (CDC) and a few state or local health departments. There is
currently no commercially available test for Zika virus.[40]

Vaccines

Vaccinations have been linked to GBS[41] by temporal association. For example, a study reviewing GBS cases during the 1992-1993 and 1993-1994 influenza seasons found an adjusted
relative risk of 1.7 cases per 1 million influenza vaccinations.[42]

In most cases, however, no definite causal relation has been established between vaccines and GBS, with the exception of rabies vaccine prepared from infected brain tissue and the 1976 swine
flu vaccine.[35, 43] (The increased risk of GBS after the swine flu vaccination, however, was just one extra case per 100,000 vaccinations.[44] )

Moreover, a review of all postvaccination cases of GBS from 1990-2005 did not reveal an increase in mortality with postvaccination cases of GBS compared with cases resulting from other
causes.[41]

In addition, some studies have called the GBS/vaccine link into question, finding no evidence of an increased risk of GBS after seasonal influenza vaccine or after the 2009 H1N1 mass
vaccination program.[45, 46, 47]

For example, a study by Kawai et al that monitored adverse events following administration of the 2012-13 influenza vaccines found no association between the vaccines and GBS. Results were
based on 3.6 million first doses of inactivated influenza vaccine in patients aged 6 months and older and 250,000 first doses of live attenuated vaccine in patients aged 2-49 years.[48]

A study by Dieleman et al researched the association between the pandemic influenza A (H1N1) 2009 vaccine and GBS in 104 patients in 5 European countries. Adjusting for the effects of
influenzalike illness/upper respiratory tract infection, seasonal influenza vaccination, and calendar time, the authors concluded that there was no increased risk of occurrence of GBS after
receiving the pandemic influenza vaccine.[49]

Similarly, a study conducted by the Chinese Centers for Disease Control found no evidence of increased risk of GBS from administration of the H1N1 vaccine, following the administration of 89.6
million doses of the vaccine between September 21, 2009 and March 21, 2010.[50]

Epidemiologic studies from Finland and southern California failed to validate an earlier retrospective study from Finland that suggested a cause-effect relationship between oral polio vaccination
and GBS.[51, 52] In contrast, a Brazilian study suggested that, based on a temporal association between the vaccine and the onset of GBS, the vaccine may rarely correlate with the syndrome.
[53]

Results from studies into the association between GBS and other vaccines include the following:

Data from a large-scale epidemiologic study reported a decreased GBS incidence following administration of tetanus toxoid containing vaccinations when compared with the baseline
population[54]

An epidemiologic study failed to show any conclusive epidemiologic association between GBS and the hepatitis B vaccine[55]

A large Latin American study of more than 2000 children with GBS following a mass measles vaccination program in 1992-1993 failed to establish a statistically significant causal
relationship between administration of the measles vaccine and GBS[56]

A report from the CDC suggests that recipients of the Menactra meningococcal conjugate vaccine may be at increased risk of GBS[57]

Case reports exist regarding group A streptococcal vaccines and the rabies vaccine; however, conclusive, statistically significant evidence is lacking

Medications

In a case-controlled study, patients with GBS reported more frequent penicillin and antimotility drug use and less frequent oral contraceptive use. However, no definite cause-effect relationships
have been established.[58]

Case reports exist in the setting of tumor necrosis factor antagonist agents used in rheumatoid arthritis.[59, 60, 61, 62] Case reports also exist regarding streptokinase, isotretinoin, danazol,
captopril, gold, and heroin, among others.

A study by Ali indicated that antibiotic therapy with fluoroquinolones also is associated with the development of GBS. Using cases reported between 1997 and 2012 to the US Food and Drug
Administration (FDA) Adverse Reporting System, he determined that of 539 reports of peripheral neuropathy associated with fluoroquinolone treatment, 9% were for patients with GBS.[63]

Other associations

Various events, such as surgery, trauma, and pregnancy, have been reported as possible triggers of GBS, but these associations remain mostly anecdotal.

Case reports cite associations between bariatric and other gastric surgeries, renal transplantation, and epidural anesthesia.[64]

Anecdotal associations include systemic lupus erythematosus, sarcoidosis, lymphoma, and snakebite.

Tumor necrosis factor–alpha polymorphisms with increased expression are associated with many autoimmune and inflammatory diseases, and may increase susceptibility to axonal GBS
subtypes. However, the role of these polymorphisms in GBS remains unclear and warrants further investigation.[65]

Epidemiology
Occurrence in the United States

The annual US incidence of GBS is 1.2-3 per 100,000 inhabitants, making GBS the most common cause of acute flaccid paralysis in the United States.[8, 1, 30, 66] In comparing age groups, the
annual mean rate of hospitalizations in the United States related to GBS increases with age, being 1.5 cases per 100,000 population in persons aged less than 15 years and peaking at 8.6 cases
per 100,000 population in persons aged 70-79 years.[67]

US military personnel are at slightly increased risk of GBS compared with the general population. An antecedent episode of infectious gastroenteritis was a significant risk factor for the
development of GBS among military personnel.[24]

https://emedicine.medscape.com/article/315632-print 4/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
International occurrence

GBS has been reported throughout the world.[68, 69] Most studies show annual incidence figures similar to those in the United States, without geographical clustering. AMAN and AMSAN occur
mainly in northern China, Japan, and Mexico, making up only 5-10% percent of GBS cases in the United States.[70] AIDP accounts for up to 90% of cases in Europe, North America, and the
developed world.

Epidemiologic studies from Japan indicate that in this region, in comparison with North America and Europe, a greater percentage of GBS cases are associated with antecedent C jejuni
infections and a lesser number are related to antecedent CMV infections. Similarly, it has been reported that 69% of GBS cases in Dhaka, Bangladesh, have clinical evidence of antecedent C
jejuni infection.[31]

Race-related demographics

GBS has been reported throughout the international community; no racial preponderance exists. In North America, Western Europe, and Australia, most patients with GBS meet
electrophysiologic criteria for demyelinating polyneuropathy. In northern China, up to 65% of patients with GBS have axonal pathology.[15]

Sex-related demographics
GBS has a male-to-female ratio of 1.5:1; male preponderance is especially seen in older patients. However, a Swedish epidemiologic study reported that GBS rates decrease during pregnancy
and increase in the months immediately following delivery.[71]

Age-related differences in incidence

GBS has been reported in all age groups, with the syndrome occurring at any time between infancy and old age. In the United States, the syndrome's age distribution seems to be bimodal, with
a first peak in young adulthood (ages 15-35 y) and a second, higher one in middle-aged and elderly persons (ages 50-75 y). Infants appear to have the lowest risk of developing GBS.[72]

Prognosis
Short of death, the worst-case scenario in GBS is tetraplegia within 24 hours, with incomplete recovery after 18 months or longer. The best-case scenario is mild difficulty walking, with recovery
within weeks. The usual scenario, however, is peak weakness in 10-14 days, with recovery in weeks to months. Average time on a ventilator (without treatment) is 50 days. There are likely many
mild cases of GBS that are never definitively diagnosed, and patients make full recovery without treatment. The spectrum of milder disease has not been well studied nor clarified.

Approximately 80% patients with GBS walk independently at 6 months, and about 60% of patients attain full recovery of motor strength by 1 year. Recovery in approximately 5-10% of patients
with GBS is prolonged, with several months of ventilator dependency and a very delayed, incomplete recovery.

Mortality

A 2008 epidemiologic study reported a 2-12% mortality rate despite ICU management,[66] although the rate may be less than 5% in tertiary care centers with a team of medical professionals
who are familiar with GBS management.

Causes of GBS-related death include acute respiratory distress syndrome (ARDS), sepsis, pneumonia, venous thromboembolic disease, and cardiac arrest. Most cases of mortality are due to
severe autonomic instability or from the complications of prolonged intubation and paralysis.[73, 74, 75, 76] The leading cause of death in elderly patients with GBS is arrhythmia.

GBS-associated mortality rates increase markedly with age. In the United States, the case-fatality ratio ranges from 0.7% in persons younger than 15 years to 8.6% in individuals older than 65
years. Survey data has shown that in patients aged 60 years or older, the risk of death is 6-fold that of persons aged 40-59 years and is 157-fold that of patients younger than 15 years. Although
the death rate increases with age in males and females, after age 40 years males have a death rate that is 1.3 times greater than that of females.

GBS-related deaths usually occur in ventilator-dependent patients, resulting from such complications as pneumonia, sepsis, acute respiratory distress syndrome, and, less frequently, autonomic
dysfunction.[77] Underlying pulmonary disease and the need for mechanical ventilation increase the risk of death, especially in elderly patients.

Morbidity

A significant percentage of survivors of GBS have persistent motor sequelae. Estimates indicate that 15-20% of patients have moderate residual deficits from GBS and that 1-10% are left
severely disabled. Although the exact prevalence is uncertain, up to 25,000-50,000 persons in the United States may have long-term functional deficits from GBS.

The speed of recovery varies. Recovery often takes place within a few weeks or months; however, if axonal degeneration has occurred, recovery can be expected to progress slowly over many
months, because regeneration may require 6-18 months. Length of hospital stay increases with advancing age, because of disease severity and associated medical complications.

Patients may experience persistent weakness, areflexia, imbalance, or sensory loss. Approximately 7-15% of patients have permanent neurologic sequelae (although figures of as high as 40%
have been estimated), including bilateral footdrop, intrinsic hand muscle wasting, sensory ataxia, and dysesthesia. Patients may also exhibit long-term differences in pain intensity, fatigability, and
functional impairment compared with healthy controls.[78, 79] In extremely rare cases, patients may experience recurrent GBS.[80, 81]

Numerous papers have addressed the issue of persistent fatigue after recovery from GBS.[82, 83, 84] Studies have suggested that a large percentage of patients continue to have fatigue-related
problems, subsequently limiting their function at home and at work, as well as during leisure activities. Treatment suggestions range from gentle exercise to improvement in sleep patterns to
relief of pain or depression, if present.

GBS can produce long-lasting changes in the psychosocial status of patients and their families.[85, 86, 87] Changes in work and leisure activities can be observed in just over one third of these
patients, and psychosocial functional health status can be impaired even years after the GBS event.

Interestingly, psychosocial performance does not seem to correlate with the severity of residual problems with physical function. Poor conditioning and easy fatigability may be contributory
factors.

Rudolph et al determined that patients who have had GBS seem overall to have a reduced quality of life and physical functioning. Their findings were based on a study of 42 GBS patients who
were examined after a median of 6 years post ̶ disease onset using a variety of measures, including the visual analogue scale (VAS) for pain, the disability rating index (DRI), and the Medical
Outcome Study 36-item short-form health status scale (SF-36).[79]

Prognostic factors

The following factors have been associated with adverse effect on outcomes in GBS[6, 88, 89] :

Preceding gastrointestinal infection or diarrheal illness

Older age (57 years or older)

Poor upper extremity muscle strength

Acute hospital stay of longer than 11 days

ICU requirement

Need for mechanical ventilation

Medical Research Council (MRC) score below 40

Discharge to rehabilitation

A rapidly progressing onset of weakness also has been associated with less favorable outcomes in many studies, although in other reports, delayed time to peak disability has been shown to be
an independent predictor of poor outcome at 1 year.

https://emedicine.medscape.com/article/315632-print 5/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
Mean compound muscle action potential (CMAP) amplitudes of less than 20% of the lower limit of normal or the presence of inexcitable nerves on initial electrophysiologic studies are other
predictors of poorer functional outcomes. Later tests (>1 mo after onset) showing persistence of a low mean CMAP have an even higher sensitivity and specificity than do initial tests showing low
amplitude.

A prospective, multicenter study by Petzold et al suggested that CSF levels of high ̶ molecular weight neurofilament (NfH) protein, an axonal protein, are prognostic indicators in GBS.[90] The
investigators found that among patients with GBS who suffered a poor outcome (defined as an inability to walk independently), the median NfH level was 1.78 ng/mL; in patients with GBS who
had a good outcome, the median level was 0.03 ng/mL.

Increased CSF levels of neuron-specific enolase and S-100b protein are also associated with longer duration of illness.[8] Serologically, a longer-lasting increase in immunoglobulin M (IgM) anti-
GM1 predicts slow recovery.[8]

The presence of underlying pulmonary disease or manifestation of dysautonomia has no prognostic significance in GBS.

A cohort study by van den Berg et al indicated that in patients with GBS, an association exists between prolonged mechanical ventilation (>2 months) and poor prognosis, although a cross-
sectional study by these investigators suggested that some patients who have undergone prolonged mechanical ventilation may demonstrate slow, persistent recovery that eventually allows
them to walk unaided and live independently. At 6 months, in the cohort study, the ability to walk unaided had been achieved in 18% of patients treated with prolonged mechanical ventilation and
in 76% of those who underwent a shorter ventilation period. However, in the cross-sectional study, which had a median follow-up period of 11 years, 58% of patients who underwent prolonged
mechanical ventilation could, at maximum follow-up, walk unaided, including 31% who achieved this outcome more than a year subsequent to diagnosis.[91]

Relapse

In a small percentage (~10%) of patients, an acute relapse occurs after initial improvement or stabilization after treatment. Some patients also demonstrate treatment fluctuations during their
clinical course. Recurrence of Guillain-Barré syndrome is rare but has been reported in 2-5% of patients.[80, 81]

There is no convincing evidence that IVIG treatment or plasma exchange has a significant effect on the rate of treatment failure or of acute relapse.[92] The risk of relapse does, however, appear
to be higher in patients in whom there has been a later onset of treatment, a more protracted disease course, and more associated medical conditions. Additional plasma exchange or IVIG
treatments often result in further improvement.[93]

Patient Education
Patients with GBS and their families should be educated on the illness, the disease process, and the anticipated course. GBS is a life event with a potentially long-lasting influence on patients'
physical and psychosocial well-being.[85, 86, 87] Family education and training also is recommended to prevent complications during the early stages of the disease and to assist in the recovery
of function during the rehabilitation stages.

For patient education information, see the Brain and Nervous System Center, as well as Guillain-Barré Syndrome.

Presentation

History
The typical patient with Guillain-Barré syndrome (GBS), which in most cases will manifest as acute inflammatory demyelinating polyradiculoneuropathy (AIDP), presents 2-4 weeks following a
relatively benign respiratory or gastrointestinal illness with complaints of finger dysesthesias and proximal muscle weakness of the lower extremities. The weakness may progress over hours to
days to involve the arms, truncal muscles, cranial nerves, and muscles of respiration. Variants of GBS may present as pure motor dysfunction or acute dysautonomia.

The mean time to the clinical function nadir is 12 days, with 98% of patients reaching a nadir by 4 weeks. A plateau phase of persistent, unchanging symptoms then ensues, followed days later
by gradual symptom improvement.[3] Progression of symptoms beyond that point brings the diagnosis under question. Recovery usually begins 2-4 weeks after the progression ceases.[94] The
mean time to clinical recovery is 200 days.

Some patients with atypical presentation, incomplete weakness, and some inconsistencies in their physical examination are frequently diagnosed as having a psychological reaction or hysteria,
and the diagnosis is very difficult. They are frequently sent home from the emergency department and then return with persistent or progressive symptoms hours to days later.

Weakness

The classic clinical picture of weakness is ascending and symmetrical in nature. The lower limbs are usually involved before the upper limbs. Proximal muscles may be involved earlier than the
more distal ones. Trunk, bulbar, and respiratory muscles can be affected as well.

Patients may be unable to stand or walk despite reasonable strength, especially when ophthalmoparesis or impaired proprioception is present. Respiratory muscle weakness with shortness of
breath may be present.

Weakness develops acutely and progresses over days to weeks. Severity may range from mild weakness to complete tetraplegia with ventilatory failure.

Cranial nerve involvement

Cranial nerve involvement is observed in 45-75% of patients with GBS. Cranial nerves III-VII and IX-XII may be affected. Common complaints include the following:

Facial droop (may mimic Bell palsy)

Diplopias

Dysarthria

Dysphagia

Ophthalmoplegia

Pupillary disturbances

Facial and oropharyngeal weakness usually appears after the trunk and limbs are affected. The Miller-Fisher variant of GBS is unique in that this subtype begins with cranial nerve deficits.[95]

Sensory changes

Most patients complain of paresthesias, numbness, or similar sensory changes. Sensory symptoms often precede the weakness. Paresthesias generally begin in the toes and fingertips,
progressing upward but generally not extending beyond the wrists or ankles. Loss of vibration, proprioception, touch, and pain distally may be present.

Sensory symptoms are usually mild. In most cases, objective findings of sensory loss tend to be minimal and variable.

Pain
In a prospective, longitudinal study of pain in patients with GBS, 89% of patients reported pain that was attributable to GBS at some time during their illness. On initial presentation, almost 50% of
patients described the pain as severe and distressing. The mechanism of pain is uncertain and may be a product of several factors. Pain can result from direct nerve injury or from the paralysis
and prolonged immobilization.

Pain is most severe in the shoulder girdle, back, buttocks, and thighs and may occur with even the slightest movements. The pain is often described as aching or throbbing in nature.

https://emedicine.medscape.com/article/315632-print 6/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
Dysesthetic symptoms are observed in approximately 50% of patients during the course of their illness. Dysesthesias frequently are described as burning, tingling, or shocklike sensations and
are often more prevalent in the lower extremities than in the upper extremities. Dysesthesias may persist indefinitely in 5-10% of patients.

Other pain syndromes in GBS include the following:

Myalgic complaints, with cramping and local muscle tenderness

Visceral pain

Pain associated with conditions of immobility (eg, pressure nerve palsies, decubitus ulcers)

The intensity of pain on admission correlates poorly with neurologic disability on admission and with the end outcome.

Autonomic changes

Autonomic nervous system involvement with dysfunction in the sympathetic and parasympathetic systems can be observed in patients with GBS.

Autonomic changes can include the following:

Tachycardia

Bradycardia

Facial flushing

Paroxysmal hypertension

Orthostatic hypotension

Anhidrosis and/or diaphoresis

Urinary retention due to urinary sphincter disturbances may be noted. Constipation due to bowel paresis and gastric dysmotility may be present. Bowel and bladder dysfunction are rarely early or
persistent findings.

A study by Anandan et al indicated that in hospitalized patients with GBS, autonomic dysfunction most frequently manifests as diarrhea/constipation (15.5%), hyponatremia (14.9%), syndrome of
inappropriate antidiuretic hormone secretion (SIADH, 4.8%), bradycardia (4.7%), and urinary retention (3.9%). The study included 2587 patients with GBS and 10,348 controls.[96]

Dysautonomia is more frequent in patients with severe weakness and respiratory failure. Autonomic changes rarely persist in a patient with GBS.

Respiratory involvement

Upon presentation, 40% of patients have respiratory or oropharyngeal weakness. Typical complaints include the following:

Dyspnea on exertion

Shortness of breath

Difficulty swallowing

Slurred speech

Ventilatory failure with required respiratory support occurs in up to one third of patients at some time during the course of their disease.

Antecedent illness

Up to two thirds of patients with GBS report an antecedent illness or event 1-3 weeks prior to the onset of weakness. Upper respiratory and gastrointestinal illnesses are the most commonly
reported conditions.[22, 24] Symptoms generally have resolved by the time the patient presents for the neurologic condition.

Campylobacter jejuni is the major causative organism identified in most studies and is responsible for cases of AIDP and acute motor axonal neuropathy (AMAN). In one major study, previous
diarrheal illness had occurred in 60% of patients with axonal GBS (by neurophysiologic testing).

Vaccinations, surgical procedures, and trauma have been reported to trigger the development of GBS.[41] Much of this information is anecdotal, although vaccination with the swine flu vaccine
administered in 1976 was shown to increase the risk of contracting GBS to a small, but definable, degree. Rabies vaccine prepared from infected brain tissue also was found to have an
association with GBS. Studies of other vaccines, however, have not shown a significant relationship between these drugs and GBS or have been inconclusive.[43, 49]

Physical Examination
Cardiac arrhythmias, including tachycardias and bradycardias, can be observed as a result of autonomic nervous system involvement. Tachypnea may be a sign of ongoing dyspnea and
progressive respiratory failure.

Blood pressure lability is another common feature, with alterations between hypertension and hypotension. Temperature may be elevated or low.

Respiratory examination may be remarkable for poor inspiratory effort or diminished breath sounds. On abdominal examination, paucity or absence of bowel sounds suggests paralytic ileus.
Suprapubic tenderness or fullness may be suggestive of urinary retention.

Neurologic examination

Facial weakness (cranial nerve VII) is observed most frequently, followed by symptoms associated with cranial nerves VI, III, XII, V, IX, and X. Involvement of facial, oropharyngeal, and ocular
muscles results in facial droop, dysphagia, dysarthria, and findings associated with disorders of the eye.

Ophthalmoparesis may be observed in up to 25% of patients with GBS. Limitation of eye movement most commonly results from a symmetrical palsy associated with cranial nerve VI. Ptosis
from cranial nerve III (oculomotor) palsy also is often associated with limited eye movements. Pupillary abnormalities, especially those accompanying ophthalmoparesis, are relatively common as
well. Papilledema secondary to elevated intracranial pressure is present in rare cases. Tonic pupils have been reported but only in severe cases.

Lower extremity weakness usually begins first and ascends symmetrically and progressively over the first several days. Upper extremity, trunk, facial, and oropharyngeal weakness is observed to
a variable extent. Marked asymmetrical weakness calls the diagnosis of GBS into question.

Patients may be unable to stand or walk despite reasonable strength, especially when ophthalmoparesis or impaired proprioception is present.

Despite frequent complaints of paresthesias, objective sensory changes are minimal. A well-demarcated sensory level should not be observed in patients with GBS; such a finding calls the
diagnosis of GBS into question.

Reflexes are absent or reduced early in the disease course. Hyporeflexia or areflexia of involved areas represents a major clinical finding on examination of the patient with GBS. Pathologic
reflexes, such as the Babinski sign, are absent. Hypotonia can be observed with significant weakness.

DDx

https://emedicine.medscape.com/article/315632-print 7/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
Diagnostic Considerations
Problems to consider in the differential diagnosis of Guillain-Barré syndrome (GBS) include the following:

Acute myelopathy (eg, from compression, transverse myelitis, vascular injury)

Chronic inflammatory demyelinating polyneuropathy

Conversion disorder/hysterical paralysis

Human immunodeficiency virus (HIV) peripheral neuropathy

Neurotoxic fish or shellfish poisoning

Paraneoplastic neuropathy

Poliomyelitis

Porphyria polyneuropathy

Spinal cord compression

Spinal cord syndromes, particularly postinfection

Tick paralysis[97]

Toxic neuropathies (eg, arsenic, thallium, organophosphates, lead)

Vasculitic neuropathies

Vitamin deficiency (eg, vitamin B-12, folate, thiamine)

West Nile encephalitis

Bilateral strokes

Acute cerebellar ataxia syndromes

Posterior fossa structural lesion

Differential Diagnoses
Botulism

Cauda Equina and Conus Medullaris Syndromes

Chronic Inflammatory Demyelinating Polyradiculoneuropathy

Emergent Management of Myasthenia Gravis

Heavy Metal Toxicity

Lyme Disease

Metabolic Myopathies

Multiple Sclerosis

Nutritional Neuropathy

Vasculitic Neuropathy

Workup

Workup

Approach Considerations
Guillain-Barré syndrome (GBS) is generally diagnosed on clinical grounds. Basic laboratory studies, such as complete blood counts (CBCs) and metabolic panels, are normal and of limited value
in the workup. They are often ordered, however, to exclude other diagnoses and to better assess functional status and prognosis. The ordering of specific tests should be guided by the patient's
history and presentation.

Electromyography (EMG) and nerve conduction studies (NCS) can be very helpful in the diagnosis. Abnormalities in NCS that are consistent with demyelination are sensitive and represent
specific findings for classic GBS. Delayed distal latencies, slowed nerve conduction velocities, temporal dispersion of waveforms, conduction block, prolonged or absent F waves, and prolonged
or absent H-reflexes are all findings that support demyelination. Needle EMG may be normal in acute nerve lesions, and it may take 3-4 weeks for fibrillation to develop. In the acute phase, the
only needle EMG abnormality may be abnormal motor recruitment, with decreased recruitment and rapid firing motor units in weak muscles. Unfortunately, electrodiagnostic studies can be
completely normal in acute GBS and a normal study does not rule GBS.[98, 99]

Frequent evaluations of pulmonary function parameters should be performed at bedside to monitor respiratory status and the need for ventilatory assistance.

Lumbar puncture for cerebrospinal fluid (CSF) studies is recommended. During the acute phase of GBS, characteristic findings on CSF analysis include albuminocytologic dissociation, which is
an elevation in CSF protein (>0.55 g/L) without an elevation in white blood cells. The increase in CSF protein is thought to reflect the widespread inflammation of the nerve roots.

Imaging studies, such as magnetic resonance imaging (MRI) and computed tomography (CT) scanning of the spine, may be more helpful in excluding other diagnoses, such as mechanical
causes of myelopathy, than in assisting in the diagnosis of GBS.

Peripheral Neuropathy Workup


A basic peripheral neuropathy workup is recommended in cases in which the diagnosis is uncertain. These studies may include the following:

Thyroid panel

Rheumatology profiles

Vitamin B-12

https://emedicine.medscape.com/article/315632-print 8/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
Folic acid

Hemoglobin A1C

Erythrocyte sedimentation rate (ESR)

Rapid protein reagent

Immunoelectrophoresis of serum protein

Tests for heavy metals

Biochemical Screening
Biochemical screening includes the following studies:

Electrolyte levels

Liver function tests (LFTs)

Creatine phosphokinase (CPK) level

ESR

The following should be considered:

LFT results are elevated in as many as one third of patients

CPK and ESR may be elevated with myopathies or systemic inflammatory conditions

A stool culture for C jejuni and a pregnancy test are also indicated

The syndrome of inappropriate antidiuretic hormone (SIADH) may occur

Serologic Studies
Serologic studies are of limited value in the diagnosis of GBS. Assays for antibodies to the following infectious agents may be considered:

C jejuni

Cytomegalovirus (CMV)

Epstein-Barr virus (EBV)

Herpes simplex virus (HSV)

HIV

Mycoplasma pneumoniae

An increase in titers for infectious agents, such as CMV, EBV, or Mycoplasma, may help in establishing etiology for epidemiologic purposes. HIV has been reported to precede GBS, and serology
should be tested in high-risk patients to establish possible infection with this agent.

Serum autoantibodies
Serum autoantibodies are not measured routinely in the workup of GBS, but results may be helpful in patients with a questionable diagnosis or a variant of GBS. Antibodies to glycolipids are
observed in the sera of 60-70% of patients with GBS during the acute phase, with gangliosides being the major target antigens.[100]

Specific antibodies found in association with GBS include the following:

Antibodies to GM1: Frequently found in the sera of patients with the acute motor axonal neuropathy (AMAN) or acute demyelinating polyradiculoneuropathy (AIDP) variants of GBS

Anti-GM1 antibodies: Elevated titers are closely associated antecedent C jejuni infections

Anti-GQ1b antibodies: Found in patients with GBS with ophthalmoplegia, including patients with the Miller-Fisher variant

Other antibodies to different major and minor gangliosides also have been found in GBS patients.

Nerve Conduction Studies


Nerve conduction studies (NCS) can be very helpful in the diagnostic workup and prognostic evaluation of patients with suspected GBS. Abnormalities in NCS that are consistent with
demyelination are sensitive and represent specific findings for classic GBS.[98]

Signs of demyelination can include the following:

Nerve conduction slowing

Prolongation of the distal latencies

Prolongation or absence of the F-waves[1, 2]

Conduction block or dispersion of responses: Evidence frequently demonstrated at sites of natural nerve compression.

Changes on NCS should be present in at least 2 nerves in regions that are not typical for those associated with compressive mononeuropathies (preferentially in anatomically distinct areas, such
as an arm and a leg or a limb and the face).

Although NCS results classically show a picture of demyelinating neuropathy in most patients, axonal neuropathy and inexcitable results are found in certain subgroups. The inexcitable studies
may represent either axonopathy or severe demyelination with distal conduction block.

Other characteristics of GBS include the following:

Nerve motor action potentials: May be decreased, but this is technically difficult to determine until the abnormality is severe; the extent of decreased action potentials correlates with
prognosis

Compound muscle action potential (CMAP): Amplitude may be decreased

https://emedicine.medscape.com/article/315632-print 9/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
Sensory abnormalities: Exhibited by most patients, but these findings are much less marked than they are in motor nerves; sural sparing is a common finding in patients with clinical
sensory deficits

Abnormal (delayed, small, or absent) H-reflex: May be noted

The needle examination is of limited value in GBS. Reduced motor unit recruitment and absent denervation help to support the suggestion of a demyelinating mechanism, although the same
changes can be observed in early axonal damage with pending wallerian degeneration. In severe cases, denervation changes may be observed later in the disease course.

In the axonal variant of the disease, absent or markedly reduced distal CMAP is observed on NCS. On needle examination, profuse and early denervation potentials (fibrillations) also support the
conclusion that there has been axonal injury.

In some cases, neurophysiologic testing is normal in patients with GBS, especially in the first 1-2 weeks of the disease. This is believed to be due to the location of demyelinating lesions in
proximal sites not amenable to study.[99] For example, a retrospective, single-center study by Luigetti et al found that in 37% of patients with GBS who underwent an early nerve conduction
study (ie, 4 days or less after disease onset), neurophysiologic results were normal. As a result, the investigators, whose study involved 71 patients with GBS, suggested that extensive
neurophysiologic assessment should be performed in patients who are in the early phases of GBS.[101]

Pulmonary Function Tests


Maximal inspiratory pressures and vital capacities are measurements of neuromuscular respiratory function and predict diaphragmatic strength. Maximal expiratory pressures also reflect
abdominal muscle strength. Frequent evaluations of these parameters should be performed at bedside to monitor respiratory status and the need for ventilatory assistance.

Forced vital capacity (FVC) is very helpful in guiding disposition and therapy.[76] Patients with an FVC of less than 15-20 mL/kg, maximum inspiratory pressure of less than 30 cm water, or a
maximum expiratory pressure of less than 40 cm water generally progress to require prophylactic intubation and mechanical ventilation. Respiratory assistance should also be considered when
there is a decrease in oxygen saturation (arterial partial pressure of oxygen [PO2] < 70 mm Hg).

Negative inspiratory force (NIF) is a relatively easy bedside test to measure respiratory muscle function and can easily be performed every half hour to hour in difficult cases. Normal is usually
greater than 60 cm water. If the NIF is dropping or nears 20 cm water, respiratory support needs to be available.

Lumbar Puncture
Most, but not all, patients with GBS have an elevated CSF protein level (>400 mg/L), with normal CSF cell counts. Elevated or rising protein levels on serial lumbar punctures and 10 or fewer
mononuclear cells/mm3 strongly support the diagnosis.

A normal CSF protein level does not rule out GBS, however, as the level may remain normal in 10% of patients. CSF protein may not rise until 1-2 weeks after the onset of weakness.

Normal CSF cell counts may not be a feature of GBS in HIV-infected patients. CSF pleocytosis is well recognized in HIV-associated GBS.

Magnetic Resonance Imaging


MRI is sensitive, but nonspecific, for diagnosis. However, it can reveal nerve root enhancement and may be an effective diagnostic adjunct.[102, 103]

Spinal nerve root enhancement with gadolinium is a nonspecific feature seen in inflammatory conditions and is caused by disruption of the blood-nerve barrier. Selective anterior nerve root
enhancement appears to be strongly suggestive of GBS,[104] with the cauda equina nerve roots being enhanced in 83% of patients.

Other Studies
Muscle biopsy may help to distinguish GBS from a primary myopathy in unclear cases. Many different abnormalities may be seen on electrocardiography, including second- and third-degree
atrioventricular (AV) block, T-wave abnormalities, ST depression, QRS widening, and various rhythm disturbances.

Histology
Lymphocyte and macrophage infiltration is observed on microscopic examination of peripheral nerves, with macrophage influx believed to be responsible for the multifocal demyelination seen in
GBS. A variable degree of wallerian degeneration also can be observed with severe inflammatory changes.

Cellular infiltrates are scattered throughout the cranial nerves, nerve roots, dorsal root ganglions, and peripheral nerves.

Treatment

Approach Considerations
Patients who are diagnosed with GBS should be admitted to a hospital for close monitoring until it has been determined that the course of the disease has reached a plateau or undergone
reversal. Although the weakness may initially be mild and nondisabling, symptoms can progress rapidly over just a few days. Continued progression may result in a neuromuscular emergency
with profound paralysis, respiratory insufficiency, and/or autonomic dysfunction with cardiovascular complications.

Approximately one third of patients require admission to an ICU, primarily because of respiratory failure. After medical stabilization, patients can be treated on a general medical/neurologic floor,
but continued vigilance remains important in preventing respiratory, cardiovascular, and other medical complications. Patients with persistent functional impairments may need to be transferred to
an inpatient rehabilitation unit.

Continued care also is needed to minimize problems related to immobility, neurogenic bowel and bladder, and pain. Early involvement of allied health staff is recommended.

Early recognition and treatment of GBS also may be important in the long-term prognosis, especially in the patient with poor clinical prognostic signs, such as older age, a rapidly progressing
course, and antecedent diarrhea.[105]

Immunomodulatory treatment has been used to hasten recovery. Intravenous immunoglobulin (IVIG) and plasma exchange have proved equally effective.

Corticosteroids (oral and intravenous) have not been found to have a clinical benefit in GBS.[106] Consequently, this class of drugs is not currently employed in treatment of the syndrome.

A few studies have investigated other medications to treat GBS; however, the trials have been small and the evidence weak,[107] highlighting the need for further investigation of potential
treatment options.

Prehospital and Emergency Department Care

https://emedicine.medscape.com/article/315632-print 10/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
Prehospital care of patients with Guillain-Barré syndrome (GBS) requires careful attention to airway, breathing, and circulation (ABCs). Administration of oxygen and assisted ventilation may be
indicated, along with establishment of intravenous access. Emergency medical services personnel should monitor for cardiac arrhythmias and transport expeditiously.

In the emergency department (ED), continuation of ABCs, intravenous treatment, oxygen, and assisted ventilation may be indicated.[108] Intubation should be performed on patients who
develop any degree of respiratory failure. Clinical indicators for intubation in the ED include the following:

Hypoxia

Rapidly declining respiratory function

Poor or weak cough

Suspected aspiration

Typically, intubation is indicated when the forced vital capacity (FVC) is less than 15 mL/kg.[109] Declining NIF to -30 cm water should cause concern and very close monitoring.[110]

Patients should be monitored closely for changes in blood pressure, heart rate, and arrhythmias. Treatment is rarely needed for tachycardia. Atropine is recommended for symptomatic
bradycardia.

Because of the lability of dysautonomia, hypertension is best treated with short-acting agents, such as a short-acting beta blocker or nitroprusside. Hypotension from dysautonomia usually
responds to intravenous fluids and supine positioning. Temporary pacing may be required for patients with second- and third-degree heart block.

Consult a neurologist if any uncertainty exists as to the diagnosis. Consult the ICU team for evaluation of need for admission to the unit.

ICU Treatment
Good supportive care is critical in the treatment of patients with GBS.[82] Admission to the ICU should be considered for all patients with labile dysautonomia, a forced vital capacity of less than
20 mL/kg, or severe bulbar palsy.[3, 4] Any patients exhibiting clinical signs of respiratory compromise to any degree also should be admitted to an ICU.[3]

Because most deaths related to GBS are associated with complications of ventilatory failure and autonomic dysfunction, many patients need to be monitored closely in ICUs by physicians
experienced in acute neuromuscular paralysis and its accompanying complications.

Competent intensive care includes the following features:

Respiratory therapy

Cardiac monitoring

Safe nutritional supplementation

Monitoring for infectious complications (eg, pneumonia, urinary tract infections, septicemia)

Respiratory therapy

Approximately one third of patients with GBS require ventilatory support. Monitoring for respiratory failure, bulbar weakness, and difficulties with swallowing help to anticipate complications.
Proper positioning of the patient to optimize lung expansion and secretion management for airway clearance is required to minimize respiratory complications.

Serial assessment of ventilatory status is needed, including measurements of vital capacity and pulse oximetric monitoring. Respiratory assistance should be considered when the expiratory vital
capacity decreases to less than 18 mL/kg or when a decrease in oxygen saturation is noted (arterial PO2< 70 mm Hg). Tracheotomy may be required in a patient with prolonged respiratory
failure, especially if mechanical ventilation is required for more than 2 weeks.

Cardiac monitoring

Close monitoring of heart rate, blood pressure, and cardiac arrhythmias allows early detection of life-threatening situations. Critically ill patients require continuous telemetry and close medical
supervision in an ICU setting.[3] Antihypertensives and vasoactive drugs should be used with caution in patients with autonomic instability. Hemodynamic changes related to autonomic
dysfunction are usually transitory, and patients rarely require long-term medications to treat blood pressure or cardiac problems.

Nutrition

Enteral or parenteral feedings are required for patients on mechanical ventilation to ensure that adequate caloric needs are met when the metabolic demand is high. Even patients who are off the
ventilator may require nutritional support if dysphagia is severe. Precautions against dysphagia and dietary manipulations should be used to prevent aspiration and subsequent pneumonias in
patients at risk.

Prevention of infection

The risk of sepsis and infection can be decreased by the use of minimal sedation, frequent physiotherapy, and mechanical ventilation with positive end-expiratory pressure where appropriate.[3]
Transfer may be appropriate if a facility does not have the proper resources to care for patients who require prolonged intubation or prolonged intensive care.

Prevention of thromboses, pressure sores, and contractures

Prevention of secondary complications of immobility is also required. Subcutaneous unfractionated or low ̶ molecular-weight heparin (LMWH) and thromboguards are often used in the treatment
of immobile patients to prevent lower-extremity deep venous thrombosis (DVT) and consequent pulmonary embolism (PE).

Prevention of pressure sores and contractures entails careful positioning, frequent postural changes, and daily range-of-motion (ROM) exercises.

Bowel and bladder management

Although bowel and bladder dysfunction is generally transitory, management of these functions is needed to prevent other complications. Initial management should be directed toward safe
evacuation and the prevention of overdistention. Monitoring for secondary infections, such as urinary tract infection, also is an area of concern. Nephropathy has been reported in pediatric
patients.[111]

Mental status management

Hospitalized patients with GBS may experience mental status changes, including hallucinations, delusions, vivid dreams, and sleep abnormalities.[112] These occurrences are thought to be
associated with autonomic dysfunction and are more frequent in patients with severe symptoms. Such problems resolve as the patient recovers. Psychiatric and psychological problems such as
depression and anxiety are likely to occur. Education, counseling, and medications are necessary to manage these problems and help the patient adjust and improve from their profound
disability.

Physical Therapy
Estimates suggest that approximately 40% of patients who are hospitalized with GBS require inpatient rehabilitation. Unfortunately, no long-term rehabilitation outcome studies have been
conducted, and treatment is often based on experiences with other neurologic conditions. The goals of the therapy programs are to reduce functional deficits and to target impairments and
disabilities resulting from GBS.

https://emedicine.medscape.com/article/315632-print 11/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
Early in the acute phase of GBS, patients may not be able to fully participate in an active therapy program. At that stage, patients benefit from daily ROM exercises and proper positioning to
prevent muscle shortening and joint contractures. Addressing upright tolerance and endurance also may be a significant issue during the early part of rehabilitation.

Active muscle strengthening can then be slowly introduced and may include isometric, isotonic, isokinetic, or progressive resistive exercises. Mobility skills, such as bed mobility, transfers, and
ambulation, are targeted functions. Patients should be monitored for hemodynamic instability and cardiac arrhythmias, especially upon initiation of the rehabilitation program. The intensity of the
exercise program also should be monitored, because overworking the muscles may, paradoxically, lead to increased weakness.

In a study by Gupta et al in 35 patients (27 with classic GBS and 8 with acute motor axonal neuropathy [AMAN]), GBS-related deficits included neuropathic pain requiring medication therapy (28
patients), foot drop necessitating ankle-foot orthosis (AFO) use (21 patients), and locomotion difficulties requiring assistive devices (30 patients). At 1-year follow-up, the authors found continued
foot drop in 12 of the AFO patients. However, significant overall functional recovery had occurred within the general cohort.[113]

Occupational and Recreational Therapy


Occupational therapy professionals should be involved early in the rehabilitation program to promote upper body strengthening, ROM, and activities that aid functional self care. Restorative and
compensatory strategies can be used to promote functional improvements. Energy conservation techniques and work simplification also may be helpful, especially if the patient demonstrates
poor strength and endurance.

Participation in recreational therapy assists in the patient's adjustment to disability and improves integration into the community. Recreational activities, either new or adapted, can be used to
promote the growth, development, and independence of a long-term hospital patient.

Speech Therapy
Speech therapy is aimed at promoting speech and safe swallowing skills for patients who have significant oropharyngeal weakness with resultant dysphagia and dysarthria. In ventilator-
dependent patients, alternative communication strategies also may need to be implemented.

Once weaned from the ventilator, patients with tracheostomies can learn voicing strategies and can eventually be weaned from the tracheostomy tube. Cognitive screening also can be
performed conjointly with neuropsychology to assess for deficits, since cognitive problems have been reported in some patients with GBS, especially those who have had an extended ICU stay.

Immunotherapy
Plasma exchange carried out over a 10-day period may aid in removing autoantibodies, immune complexes, and cytotoxic constituents from serum and has been shown to decrease recovery
time by 50%. A review of 6 randomized, controlled trials involving 649 participants found that plasma exchange helped speed recovery from GBS without causing harm, apart from being followed
by a slightly increased risk of relapse.[114]

In well-controlled clinical trials, the efficacy of IVIGs in GBS patients has been shown to equal that of plasma exchange.[115, 116, 117, 118, 119, 120, 121, 122, 123]

IVIG treatment is easier to implement and potentially safer than plasma exchange, and the use of IVIGs versus plasma exchange may be a choice of availability and convenience.[119, 124, 125]

Additionally, IVIG is the preferential treatment in hemodynamically unstable patients and in those unable to ambulate independently.[126, 127] Some evidence suggests that in select patients
who do not respond initially to IVIG, a second dose may be beneficial.[128] However, this is not currently standard therapy and warrants further investigation.

Combining plasma and IVIG has not been found to improve outcomes or shorten illness duration in GBS.[126] However, some clinicians prefer to try plasma exchange first, and if this does not
provide patient improvement then they go to IVIG. Theoretically, if IVIG is given first, then the plasma exchange will be removing the IVIG, which was just given days earlier. There are no
randomized controlled trials that allow one to decide on the best plan.

Immunotherapy for children with GBS has not been rigorously studied with randomized, well-controlled studies, but it is a standard aspect of treatment in this age group.[126, 129]
Immunotherapy for pregnant women has not been studied, and safety for use during pregnancy has not been established.

Other possible treatments modulating the immune system include complement inhibitors such as eculizumab. This has been shown to be effective in animal models of Miller-Fisher
syndrome[130, 131] and to be safe in humans.[132, 133, 134] The Japanese Eculizumab Trial for GBS (JET-GBS), a prospective, multicenter, placebo-controlled, double-blind, randomized
phase 2 study, reported eculizumab to be safe in severe cases of GBS. The investigators also found that at 24 weeks, 91.6% of patients on eculizumab could walk independently and 74% could
run, compared with 71.9% and 18%, respectively, for those on placebo. Nonetheless, the report noted that "[t]he primary outcome measure did not reach the predefined response rate."
Moreover, the study, which was designed to evaluate safety, was considered underpowered with regard to showing efficacy.[133, 134]

Corticosteroids
Corticosteroids are ineffective as monotherapy.[1, 17, 135] According to moderate-quality evidence, corticosteroids given alone do not significantly hasten recovery from GBS or affect the long-
term outcome.[135] According to low-quality evidence, oral corticosteroids delay recovery.[121, 135] Diabetes requiring insulin was significantly more common and hypertension less common
with corticosteroids.

Methylprednisolone

Substantial evidence shows that intravenous methylprednisolone alone produces neither significant benefit nor harm.[135] In combination with IVIG, intravenous methylprednisolone may hasten
recovery but does not significantly affect long-term outcome.[121, 136]

Analgesia
Pain medications may be required in inpatient and outpatient settings. A tiered pharmacologic approach that starts with nonsteroidal anti-inflammatory drugs (NSAIDs) or acetaminophen, with
narcotic agents added as needed, is usually recommended.

Narcotics should be used judiciously because patients may already be at risk for ileus.[3] Most patients do not require narcotic analgesics after the first couple of months of illness.

Adjunct medications for pain, such as tricyclic antidepressants and certain anticonvulsants, may be beneficial for dysesthetic-type pains.[3, 137] Single small, randomized, controlled trials
support the use of gabapentin or carbamazepine in the ICU for management during the acute phase of GBS.

Nonpharmacologic pain relief therapies include frequent passive limb movements, gentle massage, and frequent position changes. Desensitization techniques can be used to improve the
patient's tolerance for activities. Modalities such as transcutaneous electrical nerve stimulation (TENS) and heat may prove beneficial in the management of myalgia. Education and psychological
counseling can decrease the amount of suffering associated with this pain and disability.

Immune Adsorption
Immune adsorption is an alternative treatment for Guillain-Barré syndrome that is still in the early stages of investigation. A small, prospective study reported no difference in outcome between
patients treated with immunoadsorption and those treated with plasma exchange.[138]

https://emedicine.medscape.com/article/315632-print 12/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
In critically ill patients, a small German study reported that treatment with selective immune adsorption (SIA) seemed to be safe and effective. In comparison with treatment with SIA only,
sequential therapy with IVIG was not more effective.[139]

Prevention of Thromboembolism
Venous thromboembolism is one of the major sequelae of extremity paralysis. Time to development of DVT or pulmonary embolism varies from 4-67 days following symptom onset.[3]
Prophylaxis with gradient compression hose and subcutaneous LMWH may dramatically reduce the incidence of venous thromboembolism.[3]

True gradient compression stockings (30-40 mm Hg or higher) are highly elastic and provide compression along a gradient that is highest at the toes and gradually decreases to the level of the
thigh. This reduces capacity venous volume by approximately 70% and increases the measured velocity of blood flow in the deep veins by a factor of 5 or more.

The ubiquitous white stockings known as antiembolic stockings or thromboembolic disease (TED) hose produce a maximum compression of 18 mm Hg and rarely are fitted in such a way as to
provide adequate gradient compression. They have not been shown to be effective as prophylaxis against thromboembolism.

Consultations
Consultation with a neurologist can be helpful in the initial diagnosis, workup, and treatment of patients admitted to the medical floor with GBS.

Critical care specialists may be required for patients in the ICU to help manage respiratory failure and multiple medical complications.

Consultation with a pulmonologist may be needed to perform workup and to manage respiratory issues, such as acute respiratory distress syndrome (ARDS), pneumonia, and respiratory failure.

Consultation with a cardiologist may be required if significant cardiovascular complications, such as labile blood pressure and cardiac arrhythmias, arise from the associated autonomic
dysfunction.

Consultation with a surgeon may be required for the placement of tracheostomies, enteral feeding tubes, and central lines.

Physical medicine and rehabilitation specialists (physiatrists) should evaluate patients for impairments and disabilities arising from GBS and should help to determine the most appropriate setting
for and intensity of rehabilitation care and assist with their rehabilitation and return to function.

Long-Term Monitoring
Although follow-up studies generally have assessed patients 6-12 months after onset of GBS, some studies have reported continued improvements in strength even beyond 2 years. With
prolonged recovery possible, GBS patients with continued neurologic deficits may benefit from ongoing physical therapy and conditioning programs.

As previously mentioned, numerous papers have addressed the issue of persistent fatigue after recovery from GBS.[82, 83, 84] Studies have suggested that a large percentage of patients
continue to have fatigue-related problems, subsequently limiting their function at home and at work, as well as during leisure activities. Treatment suggestions range from gentle exercise to
improvement in sleep patterns to relief of pain or depression, if present.

GBS can produce long-lasting changes in the psychosocial status of patients and their families.[85, 86, 87] Changes in work and leisure activities can be observed in just over one third of these
patients, and psychosocial functional health status can be impaired even years after the GBS event.

Interestingly, psychosocial performance does not seem to correlate with the severity of residual problems with physical function. Poor conditioning and easy fatigability may be contributory
factors. Therefore, providing long-term attention and support for this population group is important.

Medication

Medication Summary
Immunomodulatory therapy, such as plasmapheresis or the administration of intravenous immunoglobulins (IVIGs), is frequently used in patients with Guillain-Barré syndrome (GBS).[140] The
efficacy of plasmapheresis and IVIGs appears to be about equal in shortening the average duration of disease.[115, 116, 117, 118, 141] Combined treatment has not been shown to produce a
further, statistically significant reduction in disability.

A study by Lin et al indicated that the pretreatment severity score has the strongest association with therapeutic outcome in patients with GBS who undergo double-filtration plasmapheresis, with
a higher score being linked to a poorer outcome. The study involved 60 GBS patients who underwent first-line therapy with the procedure.[142]

The decision to use immunomodulatory therapy is based on the disease's severity and rate of progression, as well as on the length of time between the condition's first symptom and its
presentation. Risks, such as thrombotic events associated with IVIG, should be taken into consideration.[143, 144] Patients with severe, rapidly progressive disease are most likely to benefit
from treatment, with faster functional recovery.[145]

Immunomodulatory Agents

Class Summary
These medications are used to improve the clinical and immunologic aspects of GBS. They may decrease autoantibody production and increase the solubilization and removal of immune
complexes.

Intravenous immunoglobulin (Bivigam, Carimune NF, Gammagard S/D, Gamunex-C, Hizentra, Octagam, Privigen)
IVIG is derived from fractionated, purified human plasma collected from a large pool of multiple donors. The product is treated with solvents and detergents to inactivate any blood-borne virus.
IVIG may work via several mechanisms, including the blockage of macrophage receptors, the inhibition of antibody production, the inhibition of complement binding, and the neutralization of
pathologic antibodies.

Low Molecular Weight Heparin

Class Summary
Low ̶ molecular-weight heparin (LMWH) is used in the prophylaxis of deep venous thrombosis (DVT). The first LMWH to become available in the United States was enoxaparin (Lovenox). LMWH
has been used widely in pregnancy, although clinical trials are not yet available to demonstrate that it is as safe as unfractionated heparin.

Reversible elevation of hepatic transaminase levels occurs occasionally. Heparin-associated thrombocytopenia has been observed with LMWH.

https://emedicine.medscape.com/article/315632-print 13/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
Enoxaparin (Lovenox)
Enoxaparin enhances the inhibition of factor Xa and thrombin by increasing antithrombin III activity. It also slightly affects thrombin and clotting time and preferentially increases the inhibition of
factor Xa.

This agent has a wide therapeutic window; the prophylactic dose is not adjusted based on the patient's weight. Enoxaparin is safer and more effective than unfractionated heparin for prophylaxis
of venous thromboembolism. The average duration of treatment is 7-14 days.

Dalteparin (Fragmin)
Dalteparin is an LMWH with antithrombotic properties. It enhances the inhibition of Factor Xa and thrombin by increasing antithrombin. It has a minimal effect on activated partial thromboplastin
time (aPTT).

Tinzaparin
Tinzaparin is an LMWH with antithrombotic properties. It enhances the inhibition of Factor Xa and thrombin by increasing antithrombin. It has a minimal effect on aPTT.

Analgesics

Class Summary
Pain medications may be required in inpatient and outpatient settings. A tiered pharmacologic approach that starts with nonsteroidal anti-inflammatory drugs (NSAIDs) or acetaminophen, with
narcotic agents added as needed, is usually recommended.

Acetaminophen (Tylenol, AspirinFree Anacin, Cetafen, Feverall, Mapap Extra Strength)


Acetaminophen is the drug of choice for the treatment of pain in patients with documented hypersensitivity to aspirin or NSAIDs, as well as in those with upper GI disease or who are taking oral
anticoagulants.

Ibuprofen (Motrin, Advil, Neoprofen, Provil)


Ibuprofen inhibits inflammatory reactions and pain by decreasing prostaglandin synthesis. It is used to provide relief of cervical myofascial pain.

Indomethacin (Indocin)
Indomethacin is used for relief of mild to moderate pain; it inhibits inflammatory reactions and pain by decreasing the activity of COX, which results in a decrease of prostaglandin synthesis.

Naproxen (Naprosyn, Naprelan, Aleve, Anaprox)


Naproxen is used for relief of mild to moderate pain; it inhibits inflammatory reactions and pain by decreasing the activity of COX, which results in a decrease of prostaglandin synthesis.

Diclofenac (Voltaren-XR, Cataflam, Zipsor, Cambia)


Diclofenac inhibits prostaglandin synthesis by decreasing COX enzyme activity, which, in turn, decreases formation of prostaglandin precursors.

Ketoprofen
Ketoprofen is used for relief of mild to moderate pain and inflammation. Small dosages are indicated initially in small patients, elderly patients, and patients with renal or liver disease. Doses
higher than 75 mg do not increase the therapeutic effects. Administer high doses with caution, and closely observe the patient's response.

Celecoxib (Celebrex)
Celecoxib primarily inhibits COX-2. COX-2 is considered an inducible isoenzyme, induced during pain and inflammatory stimuli. Inhibition of COX-1 may contribute to NSAID GI toxicity. At
therapeutic concentrations, COX-1 isoenzyme is not inhibited; thus, GI toxicity may be decreased. Seek the lowest dose of celecoxib for each patient. It is extensively metabolized in liver
primarily via cytochrome P450 2C9.

Although increased cost can be a negative factor, the incidence of costly and potentially fatal GI bleeds is clearly less with COX-2 inhibitors than with traditional NSAIDs. Ongoing analysis of cost
avoidance of GI bleeds will further define the populations that will find COX-2 inhibitors the most beneficial.

Anticonvulsants

Class Summary
Anticonvulsants may be used to alleviate painful dysesthesias, which frequently accompany peripheral neuropathies. Although they have many different mechanisms of action, their use for
alleviating neuropathic pain probably depends on their general tendency to reduce neuronal excitability.

Gabapentin (Gralise, Neurontin)


Gabapentin is a membrane stabilizer, a structural analogue of the inhibitory neurotransmitter gamma-amino butyric acid (GABA), which paradoxically is thought not to exert an effect on GABA
receptors. Gabapentin appears to exert action via the alpha(2)delta1 and alpha(2)delta2 auxiliary subunits of voltage-gaited calcium channels

It is used to manage pain and provide sedation in neuropathic pain.

Carbamazepine (Carbatrol, Equetro, Epitol, Tegretol, Tegretol-XR)


Carbamazepine is a sodium channel blocker that typically provides substantial or complete relief of pain in 80% of individuals with some forms of painful paresthesia. It reduces sustained, high-
frequency, repetitive neural firing and is a potent enzyme inducer that can induce its own metabolism. Due to potentially serious blood dyscrasias, undertake benefit-to-risk evaluation before the
drug is instituted.

https://emedicine.medscape.com/article/315632-print 14/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
Tricyclic Antidepressants

Class Summary
Tricyclic antidepressants are effective in painful paresthesias. Whereas the drugs in this category are administered in similar dosages, their sedative properties vary. Amitriptyline may be given if
the patient suffers from insomnia, whereas nortriptyline and desipramine are better choices when sedation becomes a problem.

Amitriptyline
Amitriptyline is an analgesic for certain chronic and neuropathic pain. It blocks the reuptake of norepinephrine and serotonin, which increases their concentration in the CNS. Amitriptyline
decreases pain by inhibiting spinal neurons involved in pain perception. It is highly anticholinergic. The drug is often discontinued because of somnolence and dry mouth.

Cardiac arrhythmia, especially in overdose, has been described; monitoring the QTc interval after reaching the target level is advised. Up to 1 month may be needed to obtain clinical effects.

Nortriptyline (Pamelor)
Nortriptyline has demonstrated effectiveness in the treatment of chronic pain.

By inhibiting the reuptake of serotonin and/or norepinephrine by the presynaptic neuronal membrane, this drug increases the synaptic concentration of these neurotransmitters in the CNS.

Pharmacodynamic effects such as the desensitization of adenyl cyclase and down-regulation of beta-adrenergic receptors and serotonin receptors also appear to play a role in its mechanisms of
action.

Desipramine (Norpramin)
This is the original TCA used for depression. These agents have been suggested to act by inhibiting reuptake of noradrenaline at synapses in central descending pain-modulating pathways
located in the brainstem and spinal cord.

Questions & Answers


Overview

What is Guillain-Barre syndrome (GBS)?

How does Guillain-Barre syndrome (GBS) progress?

Which cranial nerve symptoms are typical in Guillain-Barre syndrome (GBS)?

Which autonomic nervous system (ANS) symptoms may be present in Guillain-Barre syndrome (GBS)?

Which respiratory symptoms are typical in Guillain-Barre syndrome (GBS)?

How is Guillain-Barre syndrome (GBS) diagnosed?

What is the role of needle electromyography (EMG) in the diagnosis of Guillain-Barre syndrome (GBS)?

How is respiratory muscle strength evaluated in Guillain-Barre syndrome (GBS)?

What is the role of CSF protein level in the diagnosis of Guillain-Barre syndrome (GBS)?

When is admission to an ICU considered for patients with Guillain-Barre syndrome (GBS)?

What should be included in intensive care for patients with Guillain-Barre syndrome (GBS)?

When is subcutaneous unfractionated or low-molecular-weight heparin (LMWH) indicated in the treatment of Guillain-Barre syndrome (GBS)?

Which immunomodulatory agent is most effective for the treatment of Guillain-Barre syndrome (GBS)?

What is the focus of physical therapy for Guillain-Barre syndrome (GBS)?

What is the focus of occupational therapy for Guillain-Barre syndrome (GBS)?

When is speech therapy indicated for Guillain-Barre syndrome (GBS)?

What is Guillain-Barre syndrome (GBS)?

What are the common variants of Guillain-Barre syndrome (GBS)?

How is Guillain-Barre syndrome (GBS) diagnosed?

How is Guillain-Barre syndrome (GBS) usually treated?

When was Guillain-Barre syndrome (GBS) first recognized?

How does Guillain-Barre syndrome (GBS) develop?

What is the role of C jejuni infection in the pathogenesis of Guillain-Barre syndrome (GBS)?

What are pathologic findings in Guillain-Barre syndrome (GBS)?

Is there symptom overlap between the variants of Guillain-Barre syndrome (GBS)?

What are the characteristics of the acute inflammatory demyelinating polyneuropathy (AIDP) subtype of Guillain-Barre syndrome (GBS)?

What are the characteristics of the acute motor axonal neuropathy (AMAN) subtype of Guillain-Barre syndrome (GBS)?

What are the characteristics of the acute motor-sensory axonal neuropathy (AMSAN) subtype of Guillain-Barre syndrome (GBS)?

What are the characteristics of the Miller-Fisher syndrome (MFS) subtype of Guillain-Barre syndrome (GBS)?

What are the characteristics of the acute panautonomic neuropathy variant of Guillain-Barre syndrome (GBS)?

What are the characteristics of the pure sensory variant of Guillain-Barre syndrome (GBS)?

What are the characteristics of the pharyngeal-cervical-brachial variant of Guillain-Barre syndrome (GBS)?

Do combinations of Guillain-Barre syndrome (GBS) variants occur?

https://emedicine.medscape.com/article/315632-print 15/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
What is the general etiology of Guillain-Barre syndrome (GBS)?

What is the incidence of C jejuni infection in Guillain-Barre syndrome (GBS)?

What symptoms suggest C jejuni infection in Guillain-Barre syndrome (GBS)?

What is the role of C jejuni antibodies in the etiology of Guillain-Barre syndrome (GBS)?

What is the incidence of cytomegalovirus (CMV) infection in Guillain-Barre syndrome (GBS)?

What symptoms suggest cytomegalovirus (CMV) infection in Guillain-Barre syndrome (GBS)?

In addition to C jejuni and cytomegalovirus (CMV), which infectious agents are etiologic factors in Guillain-Barre syndrome (GBS)?

Does Zika virus cause Guillain-Barre syndrome (GBS)?

Which vaccines have been proven to cause Guillain-Barre syndrome (GBS)?

Do influenza vaccinations increase the risk of developing Guillain-Barre syndrome (GBS)?

Do vaccines increase or decrease the risk of developing Guillain-Barre syndrome (GBS)?

Which medications increase the risk of developing Guillain-Barre syndrome (GBS)?

What are the less common causes of Guillain-Barre syndrome (GBS)?

What is the incidence of Guillain-Barre syndrome (GBS) in the US?

What is the global incidence of Guillain-Barre syndrome (GBS)?

Does Guillain-Barre syndrome (GBS) have a racial predilection?

Is Guillain-Barre syndrome (GBS) more common in males or females?

Is Guillain-Barre syndrome (GBS) more common in certain age groups?

What is the prognosis of Guillain-Barre syndrome (GBS)?

What is the mortality rate associated with Guillain-Barre syndrome (GBS)?

What are potential causes of death in individuals with Guillain-Barre syndrome (GBS)?

Which patients are at increased risk for Guillain-Barre syndrome (GBS)-related death?

What is the prevalence of persistent motor sequelae due to Guillain-Barre syndrome (GBS)?

How quickly do sequelae of Guillain-Barre syndrome (GBS) resolve?

What are the long-term sequelae of Guillain-Barre syndrome (GBS)?

Is persistent fatigue a complication of Guillain-Barre syndrome (GBS)?

Does Guillain-Barre syndrome (GBS) negatively affect psychosocial status?

What are the negative prognostic factors of Guillain-Barre syndrome (GBS)?

Is relapse of Guillain-Barre syndrome (GBS) possible after treatment?

Which factors may increase the risk of Guillain-Barre syndrome (GBS) relapse?

What information should patients be given regarding Guillain-Barre syndrome (GBS)?

Presentation

What is the focus of the history in patients with suspected Guillain-Barre syndrome (GBS)?

What is the typical disease progression of Guillain-Barre syndrome (GBS)?

Does an atypical presentation of Guillain-Barre syndrome (GBS) increase the risk of misdiagnosis?

What is the classic presentation of weakness in Guillain-Barre syndrome (GBS)?

Which symptoms suggest cranial nerve involvement in Guillain-Barre syndrome (GBS)?

Which sensory symptoms are typically present in Guillain-Barre syndrome (GBS)?

How common is pain in Guillain-Barre syndrome (GBS)?

What is the typical presentation of pain in Guillain-Barre syndrome (GBS)?

Which autonomic nervous system (ANS) symptoms may be present in Guillain-Barre syndrome (GBS)?

What is the incidence of autonomic dysfunction in Guillain-Barre syndrome (GBS)?

What are typical respiratory symptoms in Guillain-Barre syndrome (GBS)?

What are the most common antecedent illnesses in patients with suspected Guillain-Barre syndrome (GBS)?

What are the most common etiologic triggers of Guillain-Barre syndrome (GBS)?

What are the likely physical findings in Guillain-Barre syndrome (GBS)?

How does facial weakness present in Guillain-Barre syndrome (GBS)?

How common is ophthalmoparesis in patients with Guillain-Barre syndrome (GBS)?

How does lower extremity weakness present in Guillain-Barre syndrome (GBS)?

Are sensory changes common in Guillain-Barre syndrome (GBS)?

How are reflexes affected in Guillain-Barre syndrome (GBS)?

DDX

Which disorders should be considered in the differential diagnoses of Guillain-Barre syndrome (GBS)?

https://emedicine.medscape.com/article/315632-print 16/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
What are the differential diagnoses for Guillain-Barre Syndrome?

Workup

What is the role of blood testing in the diagnosis of Guillain-Barre syndrome (GBS)?

What is the role of electromyography (EMG) and nerve conduction studies (NCS) in the diagnosis of Guillain-Barre syndrome (GBS)?

What is the role of pulmonary function monitoring in the management of Guillain-Barre syndrome (GBS)?

What is the role of CSF studies in the diagnosis of Guillain-Barre syndrome (GBS)?

What is the role of imaging studies in the diagnosis of Guillain-Barre syndrome (GBS)?

Which studies should be included in a basic peripheral neuropathy workup for Guillain-Barre syndrome (GBS)?

Which biochemical screens should be performed in the workup of suspected Guillain-Barre syndrome (GBS)?

What is the role of serologic studies in the diagnosis of Guillain-Barre syndrome (GBS)?

What is the role of serum autoantibodies testing in the diagnosis of Guillain-Barre syndrome (GBS)?

Which specific antibodies are frequently found in patients with Guillain-Barre syndrome (GBS)?

What is the purpose of nerve conduction studies in the diagnosis of Guillain-Barre syndrome (GBS)?

Which findings in nerve conduction studies indicate demyelination in patients with Guillain-Barre syndrome (GBS)?

In addition to demyelination, which nerve conductions study findings suggest Guillain-Barre syndrome (GBS)?

Is needle exam in nerve conduction studies useful in the diagnosis of Guillain-Barre syndrome (GBS)?

How often are neurophysiologic testing findings negative in Guillain-Barre syndrome (GBS)?

What is the role of maximal inspiratory pressures and vital capacities in the management of Guillain-Barre syndrome (GBS)?

What is the role of forced vital capacity (FVC) testing in the management of Guillain-Barre syndrome (GBS)?

How often should negative inspiratory force (NIF) be monitored in Guillain-Barre syndrome (GBS)?

What is the role of CSF protein level testing in Guillain-Barre syndrome (GBS)?

What is the role of MRI in the diagnosis of Guillain-Barre syndrome (GBS)?

What is the role of muscle biopsy in the diagnosis of Guillain-Barre syndrome (GBS)?

What is the role of ECG in the diagnosis of Guillain-Barre syndrome (GBS)?

Which histologic findings suggest Guillain-Barre syndrome (GBS)?

Treatment

When should patients diagnosed with Guillain-Barre syndrome (GBS) be admitted to a hospital?

What type of continued care is required for Guillain-Barre syndrome (GBS) after initial treatment?

Does early recognition and treatment improve the long-term prognosis of Guillain-Barre syndrome (GBS)?

Which immunotherapy agent is most effective in the treatment of Guillain-Barre syndrome (GBS)?

Are corticosteroids effective for the treatment of Guillain-Barre syndrome (GBS)?

What is the required prehospital care for patients with Guillain-Barre syndrome (GBS)?

How should patients with Guillain-Barre syndrome (GBS) be managed in the emergency department (ED)?

Which treatments for Guillain-Barre syndrome (GBS) are administered in the ICU?

What respiratory therapy is necessary for Guillain-Barre syndrome (GBS) in the ICU?

What is the role of cardiac monitoring for Guillain-Barre syndrome (GBS) in the ICU?

What is the role of nutrition support for Guillain-Barre syndrome (GBS) in the ICU?

How should infection be prevented in patients with Guillain-Barre syndrome (GBS) in the ICU?

How are the complications of immobility prevented in the ICU in patients with Guillain-Barre syndrome (GBS)?

How are bowel and bladder dysfunction managed in the ICU in patients with Guillain-Barre syndrome (GBS)?

How should psychiatric complications be managed in the ICU in patients with Guillain-Barre syndrome (GBS)?

Is inpatient rehabilitation required for patients with Guillain-Barre syndrome (GBS)?

How does physical therapy progress in the early acute phase of Guillain-Barre syndrome (GBS)?

Is physical recovery possible for patients with Guillain-Barre syndrome (GBS)?

What are the goals of occupational and recreational therapies in patients with Guillain-Barre syndrome (GBS)?

What is the goal of speech therapy in patients with Guillain-Barre syndrome (GBS)?

How effective is plasma exchange in the treatment of Guillain-Barre syndrome (GBS)?

When is IVIG preferred over plasma exchange in the treatment of Guillain-Barre syndrome (GBS)?

Is combining plasma and IVIG an effective treatment for Guillain-Barre syndrome (GBS)?

Is immunotherapy effective in children with Guillain-Barre syndrome (GBS)?

Is eculizumab an effective treatment for Guillain-Barre syndrome (GBS)?

Are corticosteroids effective in the treatment of Guillain-Barre syndrome (GBS)?

Is methylprednisolone effective in the treatment of Guillain-Barre syndrome (GBS)?

https://emedicine.medscape.com/article/315632-print 17/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
What is the approach to pain treatment in Guillain-Barre syndrome (GBS)?

Are narcotics used to treat pain in Guillain-Barre syndrome (GBS)?

When should adjunct medications for pain be considered in Guillain-Barre syndrome (GBS)?

Which nonpharmacologic pain relief therapies can be considered in Guillain-Barre syndrome (GBS)?

Is immune adsorption an effective treatment for Guillain-Barre syndrome (GBS)?

How is thromboembolism prevented in patients with Guillain-Barre syndrome (GBS)?

Which specialists should be consulted for Guillain-Barre syndrome (GBS)?

Is long-term follow-up indicated for Guillain-Barre syndrome (GBS)?

How is long-term fatigue managed in Guillain-Barre syndrome (GBS)?

Which long-term psychosocial changes can result from Guillain-Barre syndrome (GBS)?

Medications

Which patients are most likely to benefit from immunotherapy in Guillain-Barre syndrome (GBS)?

Which medications in the drug class Immunomodulatory Agents are used in the treatment of Guillain-Barre Syndrome?

Which medications in the drug class Low Molecular Weight Heparin are used in the treatment of Guillain-Barre Syndrome?

Which medications in the drug class Analgesics are used in the treatment of Guillain-Barre Syndrome?

Which medications in the drug class Anticonvulsants are used in the treatment of Guillain-Barre Syndrome?

Which medications in the drug class Tricyclic Antidepressants are used in the treatment of Guillain-Barre Syndrome?

Contributor Information and Disclosures

Author

Michael T Andary, MD, MS Professor, Residency Program Director, Department of Physical Medicine and Rehabilitation, Michigan State University College of Osteopathic Medicine

Michael T Andary, MD, MS is a member of the following medical societies: American Academy of Physical Medicine and Rehabilitation, American Association of Neuromuscular and
Electrodiagnostic Medicine, American Medical Association, Association of Academic Physiatrists

Disclosure: Serve(d) as a speaker or a member of a speakers bureau for: Allergan<br/>Received honoraria from Allergan for speaking and teaching. for: Allergan.

Coauthor(s)

Joyce L Oleszek, MD Assistant Professor, Department of Physical Medicine and Rehabilitation, University of Colorado at Denver Health Sciences Center, The Children's Hospital of Denver

Joyce L Oleszek, MD is a member of the following medical societies: American Academy of Physical Medicine and Rehabilitation

Disclosure: Nothing to disclose.

Kelly Maurelus, MD Clinical Assistant Professor, Department of Emergency Medicine, State University of New York Downstate Medical Center

Kelly Maurelus, MD is a member of the following medical societies: American Medical Student Association/Foundation, Student National Medical Association

Disclosure: Nothing to disclose.

Rashida Y White-McCrimmon, MD Resident Physician, Department of Emergency Medicine, Kings County Hospital Center, State University of New York Downstate Medical Center

Rashida Y White-McCrimmon, MD is a member of the following medical societies: American College of Emergency Physicians, American Medical Association, Emergency Medicine Residents'
Association

Disclosure: Nothing to disclose.

Chief Editor

Milton J Klein, DO, MBA Consulting Physiatrist, Heritage Valley Health System-Sewickley Hospital and Ohio Valley General Hospital

Milton J Klein, DO, MBA is a member of the following medical societies: American Academy of Disability Evaluating Physicians, American Academy of Medical Acupuncture, American Academy
of Osteopathy, American Academy of Physical Medicine and Rehabilitation, American Medical Association, American Osteopathic Association, American Osteopathic College of Physical
Medicine and Rehabilitation, American Pain Society, Pennsylvania Medical Society

Disclosure: Nothing to disclose.

Acknowledgements

Angela Cha-Kim, MD Assistant Professor, Director of Spinal Cord Injury, Department of Physical Medicine and Rehabilitation, Loma Linda University Medical Center

Angela Cha-Kim, MD is a member of the following medical societies: American Academy of Physical Medicine and Rehabilitation and American Paraplegia Society

Disclosure: Nothing to disclose.

Heather Rachel Davids, MD Physician, Department of Anesthesiology, Interventional Pain Medicine, University of Colorado Health Sciences Center

Heather Rachel Davids, MD is a member of the following medical societies: American Academy of Pain Medicine, American Academy of Physical Medicine and Rehabilitation, and Association of
Academic Physiatrists

Disclosure: Nothing to disclose.

Steven C Dronen, MD, FAAEM Chair, Department of Emergency Medicine, LeConte Medical Center

Steven C Dronen, MD, FAAEM is a member of the following medical societies: American Academy of Emergency Medicine and Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

J Stephen Huff, MD Associate Professor of Emergency Medicine and Neurology, Department of Emergency Medicine, University of Virginia School of Medicine

J Stephen Huff, MD is a member of the following medical societies: American Academy of Emergency Medicine, American Academy of Neurology, American College of Emergency Physicians,
and Society for Academic Emergency Medicine

https://emedicine.medscape.com/article/315632-print 18/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
Disclosure: Nothing to disclose.

Edward A Michelson, MD Associate Professor, Program Director, Department of Emergency Medicine, University Hospital Health Systems of Cleveland

Edward A Michelson, MD is a member of the following medical societies: American College of Emergency Physicians, National Association of EMS Physicians, and Society for Academic
Emergency Medicine

Disclosure: Nothing to disclose.

Andrew C Miller, MD Fellow, Department of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh Medical Center (UPMC); Attending Physician, Department of Emergency
Medicine, UPMC St Margaret's Hospital

Andrew C Miller, MD is a member of the following medical societies: American Medical Association

Disclosure: Nothing to disclose.

Aishwarya Patil, MD Physiatrist (Rehabilitation Physician), Vice Chair, Immanuel Rehabilitation Center

Aishwarya Patil, MD is a member of the following medical societies: American Academy of Physical Medicine and Rehabilitation, American Association of Neuromuscular and Electrodiagnostic
Medicine, Association of Academic Physiatrists, and Association of Physicians of India

Disclosure: Nothing to disclose.

Razi M Rashid, MD, MPH Resident Physician, Department of Neurology, Northwestern University Hospital

Disclosure: Nothing to disclose.

Daniel D Scott, MD, MA Associate Professor, Department of Physical Medicine and Rehabilitation, University of Colorado School of Medicine; Attending Physician, Department of Physical
Medicine and Rehabilitation, Denver Veterans Affairs Medical Center, Eastern Colorado Health Care System

Daniel D Scott, MD, MA is a member of the following medical societies: Alpha Omega Alpha, American Academy of Physical Medicine and Rehabilitation, American Association of
Neuromuscular and Electrodiagnostic Medicine, American Paraplegia Society, Association of Academic Physiatrists, National Multiple Sclerosis Society, and Physiatric Association of Spine,
Sports and Occupational Rehabilitation

Disclosure: Nothing to disclose.

Richard H Sinert, DO Associate Professor of Emergency Medicine, Clinical Assistant Professor of Medicine, Research Director, State University of New York College of Medicine; Consulting
Staff, Department of Emergency Medicine, Kings County Hospital Center

Richard H Sinert, DO is a member of the following medical societies: American College of Physicians and Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Medscape Salary Employment

References

1. Winer JB. Guillain Barré syndrome. Mol Pathol. 2001 Dec. 54(6):381-5. [Medline]. [Full Text].

2. Ye Y, Zhu D, Wang K, Wu J, Feng J, Ma D, et al. Clinical and electrophysiological features of the 2007 Guillain-Barré syndrome epidemic in northeast China. Muscle Nerve. 2010 Sep. 42(3):311-4.
[Medline].

3. Hughes RA, Rees JH. Clinical and epidemiologic features of Guillain-Barré syndrome. J Infect Dis. 1997 Dec. 176 Suppl 2:S92-8. [Medline].

4. Walgaard C, Lingsma HF, Ruts L, Drenthen J, van Koningsveld R, Garssen MJ, et al. Prediction of respiratory insufficiency in Guillain-Barré syndrome. Ann Neurol. 2010 Jun. 67(6):781-7. [Medline].

5. Bersano A, Carpo M, Allaria S, Franciotta D, Citterio A, Nobile-Orazio E. Long term disability and social status change after Guillain-Barré syndrome. J Neurol. 2006 Feb. 253(2):214-8. [Medline].

6. Mullings KR, Alleva JT, Hudgins TH. Rehabilitation of Guillain-Barré syndrome. Dis Mon. 2010 May. 56(5):288-92. [Medline].

7. Hughes RA, Pritchard J, Hadden RD. Pharmacological treatment other than corticosteroids, intravenous immunoglobulin and plasma exchange for Guillain-Barré syndrome. Cochrane Database Syst
Rev. 2013 Feb 28. 2:CD008630. [Medline].

8. Seneviratne U. Guillain-Barré syndrome. Postgrad Med J. 2000 Dec. 76(902):774-82. [Medline]. [Full Text].

9. Jacobs BC, Koga M, van Rijs W, Geleijns K, van Doorn PA, Willison HJ, et al. Subclass IgG to motor gangliosides related to infection and clinical course in Guillain-Barré syndrome. J Neuroimmunol.
2008 Feb. 194(1-2):181-90. [Medline].

10. Jacobs BC, van Doorn PA, Schmitz PI, Tio-Gillen AP, Herbrink P, Visser LH, et al. Campylobacter jejuni infections and anti-GM1 antibodies in Guillain-Barré syndrome. Ann Neurol. 1996 Aug. 40(2):181-
7. [Medline].

11. Koga M, Takahashi M, Masuda M, Hirata K, Yuki N. Campylobacter gene polymorphism as a determinant of clinical features of Guillain-Barré syndrome. Neurology. 2005 Nov 8. 65(9):1376-81. [Medline].

12. Kimoto K, Koga M, Odaka M, Hirata K, Takahashi M, Li J, et al. Relationship of bacterial strains to clinical syndromes of Campylobacter-associated neuropathies. Neurology. 2006 Nov 28. 67(10):1837-
43. [Medline].

13. Geleijns K, Roos A, Houwing-Duistermaat JJ, van Rijs W, Tio-Gillen AP, Laman JD, et al. Mannose-binding lectin contributes to the severity of Guillain-Barré syndrome. J Immunol. 2006 Sep 15.
177(6):4211-7. [Medline].

14. Asbury AK, Cornblath DR. Assessment of current diagnostic criteria for Guillain-Barré syndrome. Ann Neurol. 1990. 27 Suppl:S21-4. [Medline].

15. Ho TW, Mishu B, Li CY, Gao CY, Cornblath DR, Griffin JW, et al. Guillain-Barré syndrome in northern China. Relationship to Campylobacter jejuni infection and anti-glycolipid antibodies. Brain. 1995 Jun.
118 ( Pt 3):597-605. [Medline].

16. Hiraga A, Mori M, Ogawara K, Kojima S, Kanesaka T, Misawa S, et al. Recovery patterns and long term prognosis for axonal Guillain-Barré syndrome. J Neurol Neurosurg Psychiatry. 2005 May.
76(5):719-22. [Medline]. [Full Text].

17. Winer JB. Treatment of Guillain-Barré syndrome. QJM. 2002 Nov. 95(11):717-21. [Medline].

18. Brown WF, Feasby TE, Hahn AF. Electrophysiological changes in the acute "axonal" form of Guillain-Barre syndrome. Muscle Nerve. 1993 Feb. 16(2):200-5. [Medline].

19. Fisher M. An unusual variant of acute immune polyneuritis (syndrome of ophthalmoplegia, ataxia, and areflexia). N Engl J Med. 1956;255:57-65:

20. Li H, Yuan J. Miller Fisher syndrome: toward a more comprehensive understanding. Chin Med J (Engl). 2001 Mar. 114(3):235-9. [Medline].

21. Chiba A, Kusunoki S, Obata H, Machinami R, Kanazawa I. Serum anti-GQ1b IgG antibody is associated with ophthalmoplegia in Miller Fisher syndrome and Guillain-Barré syndrome: clinical and
immunohistochemical studies. Neurology. 1993 Oct. 43(10):1911-7. [Medline].

22. Baravelli M, Fantoni C, Rossi A, et al. Guillain-Barré syndrome as a neurological complication of infective endocarditis. Is it really so rare and how often do we recognise it?. Int J Cardiol. Jan 10 2008.
[Medline].

23. Jacobs BC, Rothbarth PH, van der Meché FG, Herbrink P, Schmitz PI, de Klerk MA, et al. The spectrum of antecedent infections in Guillain-Barré syndrome: a case-control study. Neurology. 1998 Oct.
51(4):1110-5. [Medline].

https://emedicine.medscape.com/article/315632-print 19/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
24. Nelson L, Gormley R, Riddle MS, Tribble DR, Porter CK. The epidemiology of Guillain-Barré Syndrome in U.S. military personnel: a case-control study. BMC Res Notes. 2009 Aug 26. 2:171. [Medline].
[Full Text].

25. van der Meché FG, Visser LH, Jacobs BC, Endtz HP, Meulstee J, van Doorn PA. Guillain-Barré syndrome: multifactorial mechanisms versus defined subgroups. J Infect Dis. 1997 Dec. 176 Suppl 2:S99-
102. [Medline].

26. Zautner AE, Johann C, Strubel A, Busse C, Tareen AM, Masanta WO, et al. Seroprevalence of campylobacteriosis and relevant post-infectious sequelae. Eur J Clin Microbiol Infect Dis. 2014 Jan 12.
[Medline].

27. Rees JH, Gregson NA, Hughes RA. Anti-ganglioside GM1 antibodies in Guillain-Barré syndrome and their relationship to Campylobacter jejuni infection. Ann Neurol. 1995 Nov. 38(5):809-16. [Medline].

28. Visser LH, van der Meché FG, Meulstee J, Rothbarth PP, Jacobs BC, Schmitz PI, et al. Cytomegalovirus infection and Guillain-Barré syndrome: the clinical, electrophysiologic, and prognostic features.
Dutch Guillain-Barré Study Group. Neurology. 1996 Sep. 47(3):668-73. [Medline].

29. Kang JH, Sheu JJ, Lin HC. Increased risk of Guillain-Barré Syndrome following recent herpes zoster: a population-based study across Taiwan. Clin Infect Dis. 2010 Sep 1. 51(5):525-30. [Medline].

30. Van Koningsveld R, Van Doorn PA. Steroids in the Guillain-Barré syndrome: is there a therapeutic window?. Neurologia. 2005 Mar. 20(2):53-7. [Medline].

31. Islam Z, Jacobs BC, van Belkum A, Mohammad QD, Islam MB, Herbrink P, et al. Axonal variant of Guillain-Barre syndrome associated with Campylobacter infection in Bangladesh. Neurology. 2010 Feb
16. 74(7):581-7. [Medline].

32. Islam Z, van Belkum A, Cody AJ, Tabor H, Jacobs BC, Talukder KA, et al. Campylobacter jejuni HS:23 and Guillain-Barre syndrome, Bangladesh. Emerg Infect Dis. 2009 Aug. 15(8):1315-7. [Medline].
[Full Text].

33. Kalra V, Chaudhry R, Dua T, Dhawan B, Sahu JK, Mridula B. Association of Campylobacter jejuni infection with childhood Guillain-Barré syndrome: a case-control study. J Child Neurol. 2009 Jun.
24(6):664-8. [Medline].

34. van den Berg B, van der Eijk AA, Pas SD, Hunter JG, Madden RG, Tio-Gillen AP, et al. Guillain-Barre syndrome associated with preceding hepatitis E virus infection. Neurology. 2014 Feb 11. 82(6):491-
7. [Medline].

35. Winer JB, Hughes RA, Anderson MJ, Jones DM, Kangro H, Watkins RP. A prospective study of acute idiopathic neuropathy. II. Antecedent events. J Neurol Neurosurg Psychiatry. 1988 May. 51(5):613-8.
[Medline]. [Full Text].

36. Zika virus. Centers for Disease Control and Prevention. Available at http://www.cdc.gov/zika/disease-qa.html. 2016 Feb 4; Accessed: 2016 Feb 8.

37. Romero S, McNeil DG Jr. Zika virus may be linked to surge in rare syndrome in Brazil. New York Times. 2016 Jan 21. Available at http://www.nytimes.com/2016/01/22/world/americas/zika-virus-may-be-
linked-to-surge-in-rare-syndrome-in-brazil.html.

38. Epidemiological update: neurological syndrome, congenital anomalies, and Zika virus infection. Pan American Health Organization/World Health Organization. Available at http://www.google.com/url?
sa=t&rct=j&q=&esrc=s&source=web&cd=20&ved=0ahUKEwjpzonp8-
jKAhXDNT4KHQERCL04ChAWCF0wCQ&url=http%3A%2F%2Fwww.paho.org%2Fhq%2Findex.php%3Foption%3Dcom_docman%26task%3Ddoc_download%26Itemid%3D%26gid%3D32879%26lang%3
2016 Jan 17; Accessed: 8 Feb 2016.

39. Barbi L, Coelho AVC, Alencar LCA, Crovella S. Prevalence of Guillain-Barré syndrome among Zika virus infected cases: a systematic review and meta-analysis. Braz J Infect Dis. 2018 Mar 12. [Medline].

40. Zika virus: diagnostic testing. Centers for Disease Control and Prevention. Available at http://www.cdc.gov/zika/hc-providers/diagnostic.html. 2016 Feb 11; Accessed: 2016 Feb 12.

41. Souayah N, Nasar A, Suri MF, Qureshi AI. Guillain-Barré syndrome after vaccination in United States: data from the Centers for Disease Control and Prevention/Food and Drug Administration Vaccine
Adverse Event Reporting System (1990-2005). J Clin Neuromuscul Dis. 2009 Sep. 11(1):1-6. [Medline].

42. Lasky T, Terracciano GJ, Magder L, Koski CL, Ballesteros M, Nash D, et al. The Guillain-Barré syndrome and the 1992-1993 and 1993-1994 influenza vaccines. N Engl J Med. 1998 Dec 17.
339(25):1797-802. [Medline].

43. Kuitwaard K, Bos-Eyssen ME, Blomkwist-Markens PH, van Doorn PA. Recurrences, vaccinations and long-term symptoms in GBS and CIDP. J Peripher Nerv Syst. 2009 Dec. 14(4):310-5. [Medline].

44. Safranek TJ, Lawrence DN, Kurland LT, et al. Reassessment of the association between Guillain-Barré syndrome and receipt of swine influenza vaccine in 1976-1977: results of a two-state study. Expert
Neurology Group. Am J Epidemiol. 1991 May 1. 133 (9):940-51. [Medline].

45. Lehmann HC, Hartung HP, Kieseier BC, Hughes RA. Guillain-Barré syndrome after exposure to influenza virus. Lancet Infect Dis. 2010 Sep. 10(9):643-51. [Medline].

46. Preliminary results: surveillance for Guillain-Barré syndrome after receipt of influenza A (H1N1) 2009 monovalent vaccine - United States, 2009-2010. MMWR Morb Mortal Wkly Rep. 2010 Jun 4.
59(21):657-61. [Medline].

47. Stowe J, Andrews N, Wise L, Miller E. Investigation of the temporal association of Guillain-Barre syndrome with influenza vaccine and influenzalike illness using the United Kingdom General Practice
Research Database. Am J Epidemiol. 2009 Feb 1. 169(3):382-8. [Medline].

48. Kawai AT, Li L, Kulldorff M, Vellozzi C, Weintraub E, Baxter R. Absence of associations between influenza vaccines and increased risks of seizures, Guillain-Barré syndrome, encephalitis, or anaphylaxis
in the 2012-2013 season. Pharmacoepidemiol Drug Saf. 2014 Feb 4. [Medline].

49. Dieleman J, Romio S, Johansen K, Weibel D, Bonhoeffer J, Sturkenboom M. Guillain-Barre syndrome and adjuvanted pandemic influenza A (H1N1) 2009 vaccine: multinational case-control study in
Europe. BMJ. 2011 Jul 12. 343:d3908. [Medline].

50. Liang XF, Li L, Liu DW, Li KL, Wu WD, Zhu BP, et al. Safety of influenza A (H1N1) vaccine in postmarketing surveillance in China. N Engl J Med. 2011 Feb 17. 364(7):638-47. [Medline].

51. Kinnunen E, Junttila O, Haukka J, Hovi T. Nationwide oral poliovirus vaccination campaign and the incidence of Guillain-Barré Syndrome. Am J Epidemiol. 1998 Jan 1. 147(1):69-73. [Medline].

52. Rantala H, Cherry JD, Shields WD, Uhari M. Epidemiology of Guillain-Barré syndrome in children: relationship of oral polio vaccine administration to occurrence. J Pediatr. 1994 Feb. 124(2):220-3.
[Medline].

53. Friedrich F. Rare adverse events associated with oral poliovirus vaccine in Brazil. Braz J Med Biol Res. 1997 Jun. 30(6):695-703. [Medline].

54. Tuttle J, Chen RT, Rantala H, Cherry JD, Rhodes PH, Hadler S. The risk of Guillain-Barré syndrome after tetanus-toxoid-containing vaccines in adults and children in the United States. Am J Public
Health. 1997 Dec. 87(12):2045-8. [Medline]. [Full Text].

55. Shaw FE Jr, Graham DJ, Guess HA, Milstien JB, Johnson JM, Schatz GC, et al. Postmarketing surveillance for neurologic adverse events reported after hepatitis B vaccination. Experience of the first
three years. Am J Epidemiol. 1988 Feb. 127(2):337-52. [Medline].

56. da Silveira CM, Salisbury DM, de Quadros CA. Measles vaccination and Guillain-Barré syndrome. Lancet. 1997 Jan 4. 349(9044):14-6. [Medline].

57. Guillain-Barré syndrome among recipients of Menactra meningococcal conjugate vaccine--United States, June-July 2005. MMWR Morb Mortal Wkly Rep. 2005 Oct 14. 54(40):1023-5. [Medline].

58. Awong IE, Dandurand KR, Keeys CA, Maung-Gyi FA. Drug-associated Guillain-Barré syndrome: a literature review. Ann Pharmacother. 1996 Feb. 30(2):173-80. [Medline].

59. Shin IS, Baer AN, Kwon HJ, Papadopoulos EJ, Siegel JN. Guillain-Barré and Miller Fisher syndromes occurring with tumor necrosis factor alpha antagonist therapy. Arthritis Rheum. 2006 May.
54(5):1429-34. [Medline].

60. Silburn S, McIvor E, McEntegart A, Wilson H. Guillain-Barré syndrome in a patient receiving anti-tumour necrosis factor alpha for rheumatoid arthritis: a case report and discussion of literature. Ann
Rheum Dis. 2008 Apr. 67(4):575-6. [Medline].

61. Kurmann PT, Van Linthoudt D, So AK. Miller-Fisher syndrome in a patient with rheumatoid arthritis treated with adalimumab. Clin Rheumatol. 2009 Jan. 28(1):93-4. [Medline].

62. Bouchra A, Benbouazza K, Hajjaj-Hassouni N. Guillain-Barre in a patient with ankylosing spondylitis secondary to ulcerative colitis on infliximab therapy. Clin Rheumatol. 2009 Jun. 28 Suppl 1:S53-5.
[Medline].

63. Ali AK. Peripheral neuropathy and Guillain-Barré syndrome risks associated with exposure to systemic fluoroquinolones: a pharmacovigilance analysis. Ann Epidemiol. 2014 Jan 2. [Medline].

64. Machado FC, Valério BC, Morgulis RN, Nunes KF, Mazzali-Verst S. Acute axonal polyneuropathy with predominant proximal involvement: an uncommon neurological complication of bariatric surgery. Arq
Neuropsiquiatr. 2006 Sep. 64(3A):609-12. [Medline].

65. Prasad KN, Nyati KK, Verma A, Rizwan A, Paliwal VK. Tumor necrosis factor-alpha polymorphisms and expression in Guillain-Barré syndrome. Hum Immunol. 2010 Sep. 71(9):905-10. [Medline].

https://emedicine.medscape.com/article/315632-print 20/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
66. Alshekhlee A, Hussain Z, Sultan B, Katirji B. Guillain-Barré syndrome: incidence and mortality rates in US hospitals. Neurology. 2008 Apr 29. 70(18):1608-13. [Medline].

67. Prevots DR, Sutter RW. Assessment of Guillain-Barré syndrome mortality and morbidity in the United States: implications for acute flaccid paralysis surveillance. J Infect Dis. 1997 Feb. 175 Suppl
1:S151-5. [Medline].

68. Kushnir M, Klein C, Pollak L, Rabey JM. Evolving pattern of Guillain-Barre syndrome in a community hospital in Israel. Acta Neurol Scand. 2008 May. 117(5):347-50. [Medline].

69. Landaverde JM, Danovaro-Holliday MC, Trumbo SP, Pacis-Tirso CL, Ruiz-Matus C. Guillain-Barré syndrome in children aged J Infect Dis</i>. 2010 Mar. 201(5):746-50. [Medline].

70. McKhann GM, Cornblath DR, Griffin JW, Ho TW, Li CY, Jiang Z, et al. Acute motor axonal neuropathy: a frequent cause of acute flaccid paralysis in China. Ann Neurol. 1993 Apr. 33(4):333-42. [Medline].

71. Jiang GX, de Pedro-Cuesta J, Strigård K, Olsson T, Link H. Pregnancy and Guillain-Barré syndrome: a nationwide register cohort study. Neuroepidemiology. 1996. 15(4):192-200. [Medline].

72. Evans OB, Vedanarayanan V. Guillain-Barré syndrome. Pediatr Rev. 1997 Jan. 18(1):10-6. [Medline].

73. Zochodne DW. Autonomic involvement in Guillain-Barré syndrome: a review. Muscle Nerve. 1994 Oct. 17(10):1145-55. [Medline].

74. Maher J, Rutledge F, Remtulla H, Parkes A, Bernardi L, Bolton CF. Neuromuscular disorders associated with failure to wean from the ventilator. Intensive Care Med. 1995 Sep. 21(9):737-43. [Medline].

75. Hund EF, Borel CO, Cornblath DR, Hanley DF, McKhann GM. Intensive management and treatment of severe Guillain-Barré syndrome. Crit Care Med. 1993 Mar. 21(3):433-46. [Medline].

76. Teitelbaum JS, Borel CO. Respiratory dysfunction in Guillain-Barré syndrome. Clin Chest Med. 1994 Dec. 15(4):705-14. [Medline].

77. Fletcher DD, Lawn ND, Wolter TD, Wijdicks EF. Long-term outcome in patients with Guillain-Barré syndrome requiring mechanical ventilation. Neurology. 2000 Jun 27. 54(12):2311-5. [Medline].

78. Dornonville de la Cour C, Jakobsen J. Residual neuropathy in long-term population-based follow-up of Guillain-Barré syndrome. Neurology. 2005 Jan 25. 64(2):246-53. [Medline].

79. Rudolph T, Larsen JP, Farbu E. The long-term functional status in patients with Guillain-Barré syndrome. Eur J Neurol. 2008 Dec. 15(12):1332-7. [Medline].

80. Das A, Kalita J, Misra UK. Recurrent Guillain Barre' syndrome. Electromyogr Clin Neurophysiol. 2004 Mar. 44(2):95-102. [Medline].

81. Roper TA, Alani SM. Recurrent Guillain-Barré syndrome: lightning does strike twice. Br J Hosp Med. 1995 Apr 19-May 2. 53(8):403-7. [Medline].

82. Hughes RA, Wijdicks EF, Benson E, Cornblath DR, Hahn AF, Meythaler JM, et al. Supportive care for patients with Guillain-Barré syndrome. Arch Neurol. 2005 Aug. 62(8):1194-8. [Medline].

83. Garssen MP, Blok JH, van Doorn PA, Visser GH. Conduction velocity distribution in neurologically well-recovered but fatigued Guillain-Barré syndrome patients. Muscle Nerve. 2006 Feb. 33(2):177-82.
[Medline].

84. de Vries JM, Hagemans ML, Bussmann JB, et al. Fatigue in neuromuscular disorders: focus on Guillain-Barré syndrome and Pompe disease. Cell. Mol Life Sci. Nov 16 2009. [Medline]. [Full Text].

85. Bernsen RA, de Jager AE, Schmitz PI, van der Meché FG. Residual physical outcome and daily living 3 to 6 years after Guillain-Barré syndrome. Neurology. 1999 Jul 22. 53(2):409-10. [Medline].

86. Bernsen RA, de Jager AE, van der Meché FG, Suurmeijer TP. How Guillain-Barre patients experience their functioning after 1 year. Acta Neurol Scand. 2005 Jul. 112(1):51-6. [Medline].

87. Bernsen RA, Jacobs HM, de Jager AE, van der Meché FG. Residual health status after Guillain-Barré syndrome. J Neurol Neurosurg Psychiatry. 1997 Jun. 62(6):637-40. [Medline]. [Full Text].

88. Khan F, Pallant JF, Ng L, Bhasker A. Factors associated with long-term functional outcomes and psychological sequelae in Guillain-Barre syndrome. J Neurol. 2010 Dec. 257(12):2024-31. [Medline].

89. Winer JB, Hughes RA, Osmond C. A prospective study of acute idiopathic neuropathy. I. Clinical features and their prognostic value. J Neurol Neurosurg Psychiatry. 1988 May. 51(5):605-12. [Medline].
[Full Text].

90. Petzold A, Brettschneider J, Jin K, Keir G, Murray NM, Hirsch NP, et al. CSF protein biomarkers for proximal axonal damage improve prognostic accuracy in the acute phase of Guillain-Barré syndrome.
Muscle Nerve. 2009 Jul. 40(1):42-9. [Medline].

91. van den Berg B, Storm EF, Garssen MJP, Blomkwist-Markens PH, Jacobs BC. Clinical outcome of Guillain-Barré syndrome after prolonged mechanical ventilation. J Neurol Neurosurg Psychiatry. 2018
Apr 7. [Medline].

92. Romano JG, Rotta FT, Potter P, Rosenfeld V, Santibanez R, Rocha B, et al. Relapses in the Guillain-Barré syndrome after treatment with intravenous immune globulin or plasma exchange. Muscle
Nerve. 1998 Oct. 21(10):1327-30. [Medline].

93. Appropriate number of plasma exchanges in Guillain-Barré syndrome. The French Cooperative Group on Plasma Exchange in Guillain-Barré Syndrome. Ann Neurol. 1997 Mar. 41(3):298-306. [Medline].

94. El Mhandi L, Calmels P, Camdessanché JP, Gautheron V, Féasson L. Muscle strength recovery in treated Guillain-Barré syndrome: a prospective study for the first 18 months after onset. Am J Phys Med
Rehabil. 2007 Sep. 86(9):716-24. [Medline].

95. Lo YL. Clinical and immunological spectrum of the Miller Fisher syndrome. Muscle Nerve. 2007 Nov. 36(5):615-27. [Medline].

96. Anandan C, Khuder SA, Koffman BM. Prevalence of Autonomic Dysfunction in Hospitalized Patients with Guillain- Barre Syndrome. Muscle Nerve. 2016 Dec 31. [Medline].

97. Diaz JH. A 60-year meta-analysis of tick paralysis in the United States: a predictable, preventable, and often misdiagnosed poisoning. J Med Toxicol. 2010 Mar. 6(1):15-21. [Medline].

98. Albers JW, Kelly JJ Jr. Acquired inflammatory demyelinating polyneuropathies: clinical and electrodiagnostic features. Muscle Nerve. 1989 Jun. 12(6):435-51. [Medline].

99. Van den Bergh PY, Piéret F. Electrodiagnostic criteria for acute and chronic inflammatory demyelinating polyradiculoneuropathy. Muscle Nerve. 2004 Apr. 29(4):565-74. [Medline].

100. Kusunoki S. Antiglycolipid antibodies in Guillain-Barré syndrome and autoimmune neuropathies. Am J Med Sci. 2000 Apr. 319(4):234-9. [Medline].

101. Luigetti M, Servidei S, Modoni A, Rossini PM, Sabatelli M, Lo Monaco M. Admission neurophysiological abnormalities in Guillain-Barré syndrome: A single-center experience. Clin Neurol Neurosurg.
2015 Aug. 135:6-10. [Medline].

102. Smith N, Pereira J, Grattan-Smith P. Investigation of suspected Guillain-Barre syndrome in childhood: What is the role for gadolinium enhanced magnetic resonance imaging of the spine?. J Paediatr
Child Health. 2010 Jul 2. [Medline].

103. Yikilmaz A, Doganay S, Gumus H, Per H, Kumandas S, Coskun A. Magnetic resonance imaging of childhood Guillain-Barre syndrome. Childs Nerv Syst. 2010 Aug. 26(8):1103-8. [Medline].

104. Perry JR, Fung A, Poon P, Bayer N. Magnetic resonance imaging of nerve root inflammation in the Guillain-Barré syndrome. Neuroradiology. 1994. 36(2):139-40. [Medline].

105. Visser LH, Schmitz PI, Meulstee J, van Doorn PA, van der Meché FG. Prognostic factors of Guillain-Barré syndrome after intravenous immunoglobulin or plasma exchange. Dutch Guillain-Barré Study
Group. Neurology. 1999 Aug 11. 53(3):598-604. [Medline].

106. Double-blind trial of intravenous methylprednisolone in Guillain-Barré syndrome. Guillain-Barré Syndrome Steroid Trial Group. Lancet. 1993 Mar 6. 341(8845):586-90. [Medline].

107. Hughes RA, Pritchard J, Hadden RD. Pharmacological treatment other than corticosteroids, intravenous immunoglobulin and plasma exchange for Guillain Barré syndrome. Cochrane Database Syst
Rev. 2011 Mar 16. CD008630. [Medline].

108. McGillicuddy DC, Walker O, Shapiro NI, Edlow JA. Guillain-Barré syndrome in the emergency department. Ann Emerg Med. 2006 Apr. 47(4):390-3. [Medline].

109. Wijdicks EF, Henderson RD, McClelland RL. Emergency intubation for respiratory failure in Guillain-Barré syndrome. Arch Neurol. 2003 Jul. 60(7):947-8. [Medline].

110. Lawn ND, Fletcher DD, Henderson RD, Wolter TD, Wijdicks EF. Anticipating mechanical ventilation in Guillain-Barré syndrome. Arch Neurol. 2001 Jun. 58(6):893-8. [Medline].

111. Ilyas M, Tolaymat A. Minimal change nephrotic syndrome with Guillain-Barré syndrome. Pediatr Nephrol. 2004 Jan. 19(1):105-6. [Medline].

112. Cochen V, Arnulf I, Demeret S, Neulat ML, Gourlet V, Drouot X, et al. Vivid dreams, hallucinations, psychosis and REM sleep in Guillain-Barré syndrome. Brain. 2005 Nov. 128:2535-45. [Medline].

113. Gupta A, Taly AB, Srivastava A, Murali T. Guillain-Barre Syndrome – rehabilitation outcome, residual deficits and requirement of lower limb orthosis for locomotion at 1 year follow-up. Disabil Rehabil.
2010. 32(23):1897-902. [Medline].

114. Raphaël JC, Chevret S, Hughes RA, Annane D. Plasma exchange for Guillain-Barré syndrome. Cochrane Database Syst Rev. 2012 Jul 11. 7:CD001798. [Medline].

115. Hadden RD, Cornblath DR, Hughes RA, Zielasek J, Hartung HP, Toyka KV, et al. Electrophysiological classification of Guillain-Barré syndrome: clinical associations and outcome. Plasma
Exchange/Sandoglobulin Guillain-Barré Syndrome Trial Group. Ann Neurol. 1998 Nov. 44(5):780-8. [Medline].

https://emedicine.medscape.com/article/315632-print 21/22
10/26/2018 https://emedicine.medscape.com/article/315632-print
116. Randomised trial of plasma exchange, intravenous immunoglobulin, and combined treatments in Guillain-Barré syndrome. Plasma Exchange/Sandoglobulin Guillain-Barré Syndrome Trial Group. Lancet.
1997 Jan 25. 349(9047):225-30. [Medline].

117. van der Meché FG, Schmitz PI. A randomized trial comparing intravenous immune globulin and plasma exchange in Guillain-Barré syndrome. Dutch Guillain-Barré Study Group. N Engl J Med. 1992 Apr
23. 326(17):1123-9. [Medline].

118. Visser LH, van der Meché FG, Meulstee J, van Doorn PA. Risk factors for treatment related clinical fluctuations in Guillain-Barré syndrome. Dutch Guillain-Barré study group. J Neurol Neurosurg
Psychiatry. 1998 Feb. 64(2):242-4. [Medline]. [Full Text].

119. Koski CL, Patterson JV. Intravenous immunoglobulin use for neurologic diseases. J Infus Nurs. 2006 May-Jun. 29(3 Suppl):S21-8. [Medline].

120. Hughes RA, Swan AV, van Doorn PA. Intravenous immunoglobulin for Guillain-Barré syndrome. Cochrane Database Syst Rev. 2010 Jun 16. CD002063. [Medline].

121. Hughes RA, Swan AV, van Koningsveld R, van Doorn PA. Corticosteroids for Guillain-Barré syndrome. Cochrane Database Syst Rev. 2006 Apr 19. CD001446. [Medline].

122. Dalakas MC. Intravenous immunoglobulin in autoimmune neuromuscular diseases. JAMA. 2004 May 19. 291(19):2367-75. [Medline].

123. Fergusson D, Hutton B, Sharma M, Tinmouth A, Wilson K, Cameron DW, et al. Use of intravenous immunoglobulin for treatment of neurologic conditions: a systematic review. Transfusion. 2005 Oct.
45(10):1640-57. [Medline].

124. Shahar E. Current therapeutic options in severe Guillain-Barré syndrome. Clin Neuropharmacol. 2006 Jan-Feb. 29(1):45-51. [Medline].

125. Bascic-Kes V, Kes P, Zavoreo I, Lisak M, Zadro L, Coric L, et al. Guidelines for the use of intravenous immunoglobulin in the treatment of neurologic diseases. Acta Clin Croat. 2012 Dec. 51(4):673-83.
[Medline].

126. Hughes RA, Wijdicks EF, Barohn R, Benson E, Cornblath DR, Hahn AF, et al. Practice parameter: immunotherapy for Guillain-Barré syndrome: report of the Quality Standards Subcommittee of the
American Academy of Neurology. Neurology. 2003 Sep 23. 61(6):736-40. [Medline].

127. Lindenbaum Y, Kissel JT, Mendell JR. Treatment approaches for Guillain-Barré syndrome and chronic inflammatory demyelinating polyradiculoneuropathy. Neurol Clin. 2001 Feb. 19(1):187-204.
[Medline].

128. van Doorn PA, Kuitwaard K, Walgaard C, van Koningsveld R, Ruts L, Jacobs BC. IVIG treatment and prognosis in Guillain-Barré syndrome. J Clin Immunol. 2010 May. 30 Suppl 1:S74-8. [Medline].

129. Yata J, Nihei K, Ohya T, Hirano Y, Momoi M, Maekawa K, et al. High-dose immunoglobulin therapy for Guillain-Barré syndrome in Japanese children. Pediatr Int. 2003 Oct. 45(5):543-9. [Medline].

130. Halstead SK, Zitman FM, Humphreys PD, Greenshields K, Verschuuren JJ, Jacobs BC, et al. Eculizumab prevents anti-ganglioside antibody-mediated neuropathy in a murine model. Brain. 2008 May.
131:1197-208. [Medline].

131. Halstead SK, Humphreys PD, Zitman FM, Hamer J, Plomp JJ, Willison HJ. C5 inhibitor rEV576 protects against neural injury in an in vitro mouse model of Miller Fisher syndrome. J Peripher Nerv Syst.
2008 Sep. 13(3):228-35. [Medline].

132. Fitzpatrick AM, Mann CA, Barry S, Brennan K, Overell JR, Willison HJ. An open label clinical trial of complement inhibition in multifocal motor neuropathy. J Peripher Nerv Syst. 2011 Jun. 16(2):84-91.
[Medline].

133. Keller DM. Eculizumab Safe for Treatment of Guillain-Barré. Medscape. 2017 Oct 4. [Full Text].

134. Misawa S, Kuwabara S, Sato Y, et al. Safety and efficacy of eculizumab in Guillain-Barré syndrome: a multicentre, double-blind, randomised phase 2 trial. Lancet Neurol. 2018 Apr 20. [Medline].

135. Hughes RA, Swan AV, van Doorn PA. Corticosteroids for Guillain-Barré syndrome. Cochrane Database Syst Rev. 2010 Feb 17. CD001446. [Medline].

136. Odaka M, Tatsumoto M, Hoshiyama E, Hirata K, Yuki N. Side effects of combined therapy of methylprednisolone and intravenous immunoglobulin in Guillain-Barré syndrome. Eur Neurol. 2005.
53(4):194-6. [Medline].

137. Pandey CK, Raza M, Tripathi M, Navkar DV, Kumar A, Singh UK. The comparative evaluation of gabapentin and carbamazepine for pain management in Guillain-Barré syndrome patients in the intensive
care unit. Anesth Analg. 2005 Jul. 101(1):220-5, table of contents. [Medline].

138. Seta T, Nagayama H, Katsura K, Hamamoto M, Araki T, Yokochi M, et al. Factors influencing outcome in Guillain-Barré Syndrome: comparison of plasma adsorption against other treatments. Clin Neurol
Neurosurg. 2005 Oct. 107(6):491-6. [Medline].

139. Galldiks N, Dohmen C, Neveling M, Fink GR, Haupt WF. Selective immune adsorption treatment of severe Guillain Barré syndrome in the intensive care unit. Neurocrit Care. 2009 Dec. 11(3):317-21.
[Medline].

140. Dalakas MC. Intravenous immunoglobulin in the treatment of autoimmune neuromuscular diseases: present status and practical therapeutic guidelines. Muscle Nerve. 1999 Nov. 22(11):1479-97.
[Medline].

141. Jayasena YA, Mudalige SP, Manchanayake GS, Dharmapala HL, Kumarasiri RP, Weerasinghe VS, et al. Physiological changes during and outcome following 'filtration' based continuous plasma
exchange in Guillain Barre Syndrome. Transfus Apher Sci. 2010 Apr. 42(2):109-13. [Medline].

142. Lin JH, Tu KH, Chang CH, et al. Prognostic factors and complication rates for double-filtration plasmapheresis in patients with Guillain-Barré syndrome. Transfus Apher Sci. 2015 Feb. 52 (1):78-83.
[Medline].

143. Brannagan TH 3rd, Nagle KJ, Lange DJ, Rowland LP. Complications of intravenous immune globulin treatment in neurologic disease. Neurology. 1996 Sep. 47(3):674-7. [Medline].

144. Paran D, Herishanu Y, Elkayam O, Shopin L, Ben-Ami R. Venous and arterial thrombosis following administration of intravenous immunoglobulins. Blood Coagul Fibrinolysis. 2005 Jul. 16(5):313-8.
[Medline].

145. van Doorn PA. What's new in Guillain-Barré syndrome in 2007-2008?. J Peripher Nerv Syst. 2009 Jun. 14(2):72-4. [Medline].

146. van Doorn PA. Diagnosis, treatment and prognosis of Guillain-Barré syndrome (GBS). Presse Med. 2013 Jun. 42(6 Pt 2):e193-201. [Medline].

https://emedicine.medscape.com/article/315632-print 22/22

Potrebbero piacerti anche