Guillain Barre Review
Guillain Barre Review
DOI: 10.1111/ene.16365
REVIEW ARTICLE
KEYWORDS
diagnosis, Guillain-Barré syndrome, immunopathology, prognosis, treatment
This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2024 The Author(s). European Journal of Neurology published by John Wiley & Sons Ltd on behalf of European Academy of Neurology.
14681331, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ene.16365 by Cochrane Mexico, Wiley Online Library on [07/06/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2 of 15 BELLANTI and RINALDI
E PI D E M I O LO G Y Disease initiation
There are 100,000 new cases of GBS every year [3]. Estimates of Prodromal infections
yearly incidence (per 100,000 people) are lowest in Japan (0.44)
[4], China (0.67) [5], Tanzania (0.83) [6], and Finland (0.84) [7], In most cases, GBS is a postinfectious disease, and two-thirds of pa-
and highest in Chile (2.12) [8] and Bangladesh (3.25) [9], likely tients report prodromal gastrointestinal or respiratory symptoms.
due to differences in exposure to infectious organisms. Seasonal Campylobacter jejuni, the most commonly identified pathogenic trigger,
variations are described [10], and spikes of GBS have been re- leads to GBS in approximately 1 out of every 1000 cases, through mo-
ported following infectious outbreaks, most notably in relation lecular mimicry between its surface lipo-oligosaccharide (LOS) and host
to Campylobacter jejuni [11] and Zika virus (ZIKV) [12, 13]. Older peripheral nerve gangliosides (Figure 1). This triggers the production of
people are more commonly affected (peak incidence of GBS is cross-reactive antibodies targeting gangliosides like GM1, GD1a, and
between 50 and 70 years of age) and the male:female ratio is 1.5:1 GQ1b, resulting in axoglial damage. Other pathogens associated with
[3, 14, 15]. GBS include Epstein–Barr virus (EBV), cytomegalovirus (CMV), hepatitis
E virus (HEV), Mycoplasma pneumoniae, Haemophilus influenzae, influ-
enza A virus, and Zika virus. Mycoplasma pneumoniae is associated with
PATH O PH YS I O LO G Y O F G B S anti-galactocerebroside antibodies of the IgG isotype, more frequently
in children [16, 17]. The mechanistic association between most of these
The pathophysiology of GBS can be delineated into two pivotal pathogens and axoglial damage remains unclear, and molecular mimicry
stages: initiation by an immunological trigger and immune-mediated between C. jejuni and peripheral nerve glycans is still the best explained
disruption of axons and/or myelin. Based on electrophysiology, GBS postinfectious link in GBS, although how patients break tolerance to self-
has been traditionally divided into two forms: acute inflammatory glycans following C. jejuni infection has not been clarified.
demyelinating polyradiculoneuropathy (AIDP) and acute motor ax-
onal neuropathy (AMAN). The notion that this neurophysiological
dichotomy reflects a true underlying pathological difference be- Vaccines associated with GBS
tween primarily demyelinating versus axonal GBS is currently being
challenged, and the new EAN/PNS guidelines no longer support the Epidemiological evidence links some vaccines with a subsequent di-
distinction between AIDP and AMAN. agnosis of GBS. These include the vaccine for the ‘swine flu’ (A/New
F I G U R E 1 Lipo-oligosaccharide (LOS) on the outer membrane of Campylobacter jejuni induces cross-reactive antibodies which, through
molecular mimicry, bind to the structurally identical glycans (areas in green) present on peripheral nerve gangliosides (GM1 and GQ1b in the
example above), resulting in damage to axons and Schwann cells.
|
14681331, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ene.16365 by Cochrane Mexico, Wiley Online Library on [07/06/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GUILLAIN-BARRÉ SYNDROME: A REVIEW 3 of 15
Jersey/76 influenza), the recombinant zoster vaccine (RZV), and the antibody-mediated primary Schwann cell damage occurs, and the
adenovirus-
vector SARS-
CoV-
2 vaccines. The ‘swine flu’ vaccine potential contribution of T cells to B cell activation, remains to be
campaign in the USA was associated with 4.9–5.9 GBS cases per definitively established.
million vaccinations [18]. Though initial concerns were raised about Animal models [33–38] using active immunisation or passive
the H1N1 influenza A vaccination in 2009, subsequent surveillance transfer have shown that axoglial damage can be mediated by gan-
reported only 1–1.6 cases per million doses [19–21]. Similarly, her- glioside antibodies. In these models, autoantibodies target axonal
pes zoster vaccination with RZV showed a marginally increased risk or glial antigens and cause damage through complement activation
of GBS, with approximately 3 excess cases per million vaccinations and formation of the membrane attack complex (MAC) or induce
[22]. Adenovirus-vectored SARS-CoV-2 vaccines, such as ChAdOx1 non-complement-mediated effects on myelin and axonal regen-
and Janssen COVID-19 vaccines, have been linked to around 5.7 ex- eration. Complement-
dependent membrane disruption leads to
cess GBS cases per million first doses [23–25]. The 2023 EAN/PNS aberrant influx of calcium into the axon, calpain activation, and ulti-
GBS guidelines conclude that the advantages of vaccination (reduc- mately damage to the node of Ranvier, the paranode, and the motor
tion in morbidity and mortality related to infection and infection- nerve terminals (Figure 2a) [35, 36, 39, 40]. Ganglioside antibody
associated GBS) significantly outweigh any marginal elevation in the binding also leads to displacement of voltage-gated sodium chan-
risk of post-vaccine GBS [2]. It remains uncertain whether repeat or nels, cytoskeletal anchoring proteins, and cell adhesion molecules
future vaccination is safe following presumed vaccine-associated within the nodal and paranodal regions [38]. In transgenic mice
GBS, and certain individuals may be more susceptible to autoinflam- where GM1 is only expressed by Schwann cells and not axons, GM1
mation due to genetic predisposition. The risk of post-vaccine GBS antibodies cause primary glial injury. However, following Schwann
should be acknowledged and balanced against the benefits of vac- cell/myelin dehiscence, axonal nanoruptures are formed, ultimately
cination overall. resulting in a convergence of the pathological process with influx
of calcium, calpain activation within the axon, and cleavage of cy-
toskeletal and transmembrane proteins [41]. Such findings suggest
Other proposed or established causal associations that regardless of whether the initial injury is axonal or glial, there
is a common final pathway which leads to axonal disruption and
The risk of developing GBS may be higher during the first 6 weeks functional failure.
after surgery [26], most significantly orthopaedic or gastrointestinal Ultimately, nerve conduction failure and the related spectrum of
[27], and particularly in the context of active malignancy [28]. Cases neurological symptoms seen in patients with GBS are due to dam-
of GBS in stem cell transplant patients immunosuppressed with tac- age to neurons (node, paranode, juxtaparanode, terminal axons) or
rolimus for graft versus host disease (GVHD) prophylaxis have been glia (myelin, Schwann cells) or both. These alterations then lead to
reported [29], as well as in patients treated with immune checkpoint reversible conduction failure with the potential for rapid recovery,
inhibitors [30]; however, the actual risk has not been ascertained in demyelination with slower recovery, or axonal degeneration with
controlled studies. protracted and more often incomplete recovery, or a combination of
all three within the same nerve [38].
At least 40% of GBS patients do not have identifiable serum or
Immune-mediated axoglial disruption CSF autoantibodies. This may be because the full range of relevant
antigenic targets have yet to be found. Glycolipid domains can as-
The inflammatory nature of GBS was first acknowledged in the late sociate to form complex hetero-dimeric clusters, and around half
1940s, when prominent lymphocyte infiltration was found in post- of GBS patients have antibodies targeting these complexes but not
mortem nerve biopsies [31]. It is now clear that both innate and cel- their individual components in isolation [42, 43]. Furthermore, the
lular immune mechanisms are implicated in GBS, and T cells, B cells, pathogenic potential of certain antibodies is determined by local in-
NK cells, dendritic cells, and macrophages are all likely to contribute teraction of glycolipids with neighbouring gangliosides, which can
to axonal damage and demyelination. prevent binding of autoantibodies.
A recent study investigating autoreactive T cell immunity in pa- Genetic factors might also play a role in the immunobiology of
tients with GBS [32] found that patients with AIDP had CD4+ and GBS. A metanalysis found a moderate association between GBS and
CD8+ T cells in their blood, cerebrospinal fluid (CSF) and nerve tis- a tumour necrosis factor (TNF) gene polymorphism [44]. An associa-
sue reactive to myelin protein 0 (P0), myelin protein 2 (P2), or periph- tion has also been found between GBS severity and polymorphism of
eral myelin protein 22 (PMP22) (Figure 2b). Autoreactive memory the mannose-binding lectin (MBL) gene [45], which contributes to the
CD4+ T cells showed a proinflammatory cytotoxic TH1-like pheno- activation of the complement system and to the clearance of apop-
type and expressed genes with a known autoimmunity association. totic cells by macrophages and dendritic cells. Genetic studies on large
A similar T cell autoreactivity was not found in patients with AMAN, GBS cohorts have yet to be conducted, and genome-wide association
suggesting that patients with demyelinating and axonal GBS may studies in large, deeply phenotyped cohorts are needed to determine
actually have distinct immunopathological mechanisms. Whether whether and to what extent GBS is also genetically determined.
|
14681331, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ene.16365 by Cochrane Mexico, Wiley Online Library on [07/06/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
4 of 15 BELLANTI and RINALDI
F I G U R E 2 (a) T cells reactive to peripheral nerve myelin antigens. A humoral mechanism underlying primary glial damage has long been
hypothesised, but anti-myelin antibodies have yet to be identified. It remains unclear whether T cells are activated by macrophages (or other
antigen-presenting cells [APCs]), or by the Schwann cells themselves. (b) Ganglioside antibody binding leads to complement activation,
membrane attack complex (MAC) formation, calcium influx into the axon, calpain-mediated cytoskeletal disruption, and ultimately damage
to the node of Ranvier, the paranode, and the motor nerve terminals. Dendritic cells, macrophages, and other phagocytes are also typically
found within the nerve in patients with Guillain-Barré syndrome.
|
14681331, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ene.16365 by Cochrane Mexico, Wiley Online Library on [07/06/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GUILLAIN-BARRÉ SYNDROME: A REVIEW 5 of 15
Autonomic dysfunction in the first week within onset of symptoms, reduced sensory nerve
action potentials (SNAP) and/or CMAP would support a diagnosis
Recent gastrointestinal or respiratory infection (<6 weeks)
of peripheral neuropathy, and the finding of absent H-reflexes may
Back pain (interscapular or radicular)
suggest a radiculopathy. H-reflexes are the electrophysiological cor-
Other supportive findings
relate of reflex muscle activation after electrical stimulation of affer-
Cerebrospinal fluid albuminocytological dissociation
ent sensory fibres; their absence is highly sensitive (95%–100%) [49,
Electrophysiology confirming peripheral neuropathy
50] for GBS and their presence makes a diagnosis of GBS unlikely
|
14681331, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ene.16365 by Cochrane Mexico, Wiley Online Library on [07/06/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
6 of 15 BELLANTI and RINALDI
F I G U R E 3 Clinical features that support or refute a diagnosis of Guillain-Barré syndrome (GBS). CNS, central nervous system; CSF,
cerebrospinal fluid.
[51, 52]. The finding of a sural-sparing sensory pattern also facili- inflammatory, and monophasic. Mimics include peripheral and cen-
tates early diagnosis of GBS and aids differential diagnosis with mim- tral nervous system disorders, and in some cases only time will
ics [53, 54]. confirm or refute a diagnosis of GBS. Figure 4 summarises the dif-
ferential diagnosis of GBS based on localisation.
Imaging
Peripheral nerve mimics of GBS
Magnetic resonance imaging (MRI) and ultrasound (USS) are not
used as routine tests for the diagnosis of GBS in typical presenta- CIDP may have a GBS-like acute presentation with sensory-motor
tions but may be considered in cases where the diagnosis is uncer- disturbance (acute CIDP or A-CIDP). Similarly, autoimmune nodopa-
tain. For example, MRI of the spinal cord might help distinguish a thies (AIN) can be acute and monophasic, but often have additional
peripheral neuropathy from a myelopathy or might localise the pa- features. In particular, patients with NF186 or pan-
neurofascin
thology to the nerve roots. It should be noted, however, that diffuse antibodies (against NF155, NF140, and NF186) often have a se-
nerve thickening on MRI is non-specific and can indicate a wide dif- vere, GBS-like neuropathy with cranio-respiratory and autonomic
ferential of neuropathies including inflammatory, infective, infiltra- involvement. Three or more TRFs, progression or (re)deterioration
tive, and genetic. over 8 weeks from onset, or substantial sensory disturbance (in-
cluding sensory ataxia) make A-CIDP or an autoimmune nodopathy
more likely. Slow disease onset (defined as >2 weeks from onset to
D I FFE R E NTI A L D I AG N OS I S O F G B S nadir) [2] and/or significant early reduction in motor CV may also
suggest an alternative diagnosis. It is important to distinguish GBS
An inflammatory neuropathy is likely if onset and progression are from CIDP as the latter can respond to corticosteroids, often re-
acute or subacute (over days or weeks), along with elevated CSF quires long-term or repeated treatment, and might be responsive to
protein and improvement of symptoms upon immunosuppression or steroid-sparing immunosuppression. On the contrary, autoimmune
immunomodulation. GBS is most likely if the neuropathy is acute, nodopathies respond poorly to conventional therapies (IVIg, PLEX,
|
14681331, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ene.16365 by Cochrane Mexico, Wiley Online Library on [07/06/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GUILLAIN-BARRÉ SYNDROME: A REVIEW 7 of 15
TA B L E 2 The Guillain-Barré syndrome Typical GBS Purely motor or sensory-motor GBS, with symmetrical
spectrum. weakness of arms and legs and reduced/absent deep
tendon reflexes
GBS variants
Pharyngeal-cervical-brachial Bilateral facial weakness ± bilateral arm
paraesthesias ± weakness of the arms (no involvement of
lower limbs or cranial nerves)
Paraparetic Isolated involvement of both lower limbs
Pure sensory Sensory symptoms and reduced/absent reflexes but no
weakness
MFS spectrum (GQ1b antibody syndromes)
MFS Ataxia, ophthalmoplegia, areflexia – the full triad is present
90% of patients have anti-GQ1b antibodies
Partial MFS Two or one out of the three clinical features of MFS
Anti-GQ1b antibodies found in similar percentage to MFS
GBS/MFS overlap syndrome Features of both GBS and MFS
Around 70% of patients have anti-GQ1b antibodies
Bickerstaff brainstem Ataxia, ophthalmoplegia, hyperreflexia, altered level of
encephalitis consciousness
66% of patients have anti-GQ1b antibodies
Acute ophthalmoplegia Acute paralysis of the external (extraocular) and/or
(without ataxia) internal (intrinsic) eye muscles without ataxia. Unilateral
involvement may occur [93]
TA B L E 3 Ganglioside antibodies in
Ganglioside antibody Associated clinical findings and utility
Guillain-Barré syndrome and associated
clinical utility. Anti-GM1 Elevated IgG and IgM at baseline and persistently high
IgG early in the disease course correlate with poor clinical
recovery [94]
Also present in up to 80% of patients with MMN which
presents with weakness (usually in the upper limbs) and
conduction block on electrophysiology. This is important as
MMN does not respond to plasma exchange
Anti-GM2 May suggest CMV infection
Anti-GD1a Younger patients with facial weakness and prominent axonal
damage [95]
Associated with poor outcome [96]
Anti-GT1a Often indicate bulbar involvement in the pharyngeal-cervical-
brachial variant of GBS [97]
Anti-GQ1b Highly specific for MFS, BBE, and GBS/MFS overlap syndrome
corticosteroids) and may benefit from B cell depleting immunomod- lower back, interscapular or radicular, and may then persist for several
ulation with the monoclonal antibody rituximab if given early in the months, unresponsive to immunosuppression [58]. Infective peripheral
disease course [55–57]. neuropathies that can resemble GBS and present with acute flaccid
Other peripheral nerve disorders may mimic GBS. Vasculitic neu- paralysis include those caused by Borrelia burgdorferi (Lyme disease),
ropathies can be acute (although more frequently subacute, with HIV, pure paralytic forms of rabies, and diphtheric polyneuropathy.
onset over several weeks and progression over months) but are usually Poliovirus tends to affect the anterior horn cells of the spinal cord,
asymmetrical and multifocal (mononeuritis multiplex) whereas GBS, and poliomyelitis-related neuronopathy occurs in unvaccinated indi-
even in its variant forms (except rare monomelic cases), is symmetri- viduals, often after a prodromal flu-like phase with fever and myalgia
cal. In the early phase of disease, GBS pain is most commonly in the [59, 60]. Non-polio enteroviruses, instead, typically cause diarrhoea,
|
14681331, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ene.16365 by Cochrane Mexico, Wiley Online Library on [07/06/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
8 of 15 BELLANTI and RINALDI
F I G U R E 4 Differential diagnosis of Guillain-Barré syndrome. CIDP, chronic inflammatory demyelinating polyradiculoneuropathy; CNS,
central nervous system; LEMS, Lambert–Eaton myasthenic syndrome; MOGAD, myelin oligodendrocyte glycoprotein antibody-associated
disease.
fever and general malaise followed, 1 or 2 weeks later, in around 20% Neuromuscular junction disorders
of cases, by peripheral neuropathy [61]. HIV-related neuropathy is
usually due to seroconversion and may mimic GBS, but CSF WCC is Myasthenia gravis (MG), the most common neuromuscular junction
typically high. HIV patients with low CD4 count and severe immuno- (NMJ) disorder, causes fatigable weakness with diurnal fluctua-
depression can develop neuropathy also from opportunistic infections, tions, preserved reflexes, and decremental response to repetitive
and responsible pathogens include CMV, EBV, herpes simplex virus, nerve stimulation on electrophysiology. Bulbar-onset MG and MG
and varicella-zoster virus. Rarely, HIV patients develop weakness due with positive anti-
muscle-
specific kinase (anti-
MuSK) antibodies
to antiretroviral therapy or vitamin B12 deficiency [60, 62]. Poisoning may mimic the pharyngeal-
cervical-
brachial variant of GBS (and
with heavy metals (thallium, lead, arsenic, mercury) may cause a sub- vice versa). Lambert–Eaton myasthenic syndrome (LEMS) is often
acute or acute peripheral neuropathy mimicking GBS. In some cases, paraneoplastic, causing weakness that improves with exercise, loss
characteristic features such as upper limb weakness and wrist drop of reflexes, and incremental response to repetitive nerve stimula-
(lead) or pronounced sensory symptoms with ataxia (mercury) help the tion. Although MG, LEMS, and GBS all cause muscle weakness, di-
distinction from GBS. Similarly to GBS, peripheral neuropathy due to urnal fluctuations and exercise-induced effects are not typical of
vitamin B1 (thiamine) deficiency may present with areflexia and pro- GBS, where reflexes are characteristically absent. Botulism, another
gressive ascending weakness, but does not cause albuminocytological NMJ disorder, presents with acute paralysis and can mimic GBS,
dissociation and does not have demyelinating features on nerve con- with distinct features including descending paralysis and mydriasis.
duction studies. Finally, acute intermittent porphyria (AIP) can cause a Organophosphate intoxication causes flaccid paralysis with a constel-
GBS-like neuropathy which usually starts with symmetrical, proximal lation of cholinergic symptoms such as diarrhoea, increased urinary
weakness in the upper limbs, followed by involvement of the lower frequency, lacrimation, excessive sweating and salivation, with myo-
limbs. Less commonly, weakness begins distally with wrist or foot drop, sis and bradycardia.
and sensory disturbance is unusual [63]. Porphyric neuropathy can
progress rapidly to tetraparesis, respiratory failure, and death if un-
treated, and tends to follow a prodromal phase with neuropsychiatric Myelopathies
symptoms, abdominal pain, nausea, vomiting, and dysautonomia. Both
AIP-related neuropathy and GBS have acute onset, rapid progression, Acute spinal cord disorders can mimic GBS in the first few days
and frequent autonomic disturbance; however, porphyric neuropathy following onset. Both acute transverse myelitis (inflammatory or
tends to be axonal on electrophysiology, lacks albuminocytological dis- infective) and GBS can present with acute flaccid paraparesis,
sociation on CSF, and is unresponsive to immunotherapy. sphincter disturbance with or without back pain, and reflexes
|
14681331, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ene.16365 by Cochrane Mexico, Wiley Online Library on [07/06/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GUILLAIN-BARRÉ SYNDROME: A REVIEW 9 of 15
can be absent in early myelopathies. Myelin oligodendrocyte using the Medical Research Council sum score (MRC-SS), the GBS
glycoprotein-
associated disease (MOGAD) may present with Disability Scale (GDS), formerly known as Hughes Disability Score,
conus medullaris syndrome mimicking GBS [64], usually with the Inflammatory Rasch-built Overall Disability Scale (I-RODS), the
prominent sphincter disturbance at onset which is uncommon in Inflammatory Neuropathy Cause and Treatment (INCAT) disability
GBS. However, a cord disorder will have a sensory level whereas scale, and the Overall Neuropathy Limitation Scale (ONLS). Details
GBS will usually not, and MRI (and serology where appropriate) of the GBS outcome measures can be found in Appendix S1.
will aid differential diagnosis.
FLU I D B I O M A R K E R S I N G B S
Myopathies
The introduction of single molecule array technology has had a pro-
Acute muscle disorders include hypokalaemic periodic paralysis, some found impact on biomarker discovery, and there is mounting interest
forms of acute myositis secondary to viral infections, and acute col- in serum and CSF biomarkers of peripheral nerve disease. Three bio-
chicine myopathy. Hypokalaemic periodic paralysis causes recurrent markers have been shown to be potentially useful in GBS: neurofila-
episodes of flaccid paralysis, while viral myositides, like those from ment light chain (NfL), peripherin, and total tau (T-t au).
influenza A, lead to rapid paralysis with myalgia, rhabdomyolysis, In patients with GBS, median serum levels of NfL on admission
and fever, and may be preceded by prodromal flu-like symptoms. are significantly higher compared to age-matched healthy controls
Unlike GBS, hypokalaemic paralysis lacks sensory deficits, sphinc- and return to physiological levels 1 year after disease onset [66].
ter disturbance and bulbar or respiratory muscle involvement. High serum NfL correlates with “axonal” neurophysiology and dis-
Rhabdomyolysis and fever are common in viral myositides but rare ease severity, and high baseline levels are associated with inability
in GBS. Acute colchicine myopathy often manifests with rapidly to run and lower I-RODS at 1 year. NfL might be used to improve
progressive weakness preceded by diarrhoea; however, the lack of current outcome measures and prognostic models in GBS; however,
cranial nerve involvement, raised creatine kinase, and the finding of it is highly non-specific and elevated in at least 80 peripheral and
myotonic discharges on electromyography help in differentiating it central nervous system (CNS) diseases [67].
from GBS [65]. Serum levels of peripherin, a type III intermediate filament
protein, have been shown to be significantly elevated in GBS pa-
tients with electrophysiological evidence of axonal damage [68].
Higher central nervous system disorders Peripherin can differentiate acute nodo-p aranodal axonal injury
from slowly progressive demyelination, and discriminates cen-
Brainstem stroke may mimic GBS, especially if pontine or mesence- tral versus peripheral NfL elevations by selectively rising in GBS
phalic. Pontine strokes due to basilar artery occlusion (or dissection- and not in CNS disorders such as multiple sclerosis and dementia.
related subarachnoid haemorrhage or pontine compression) cause Further work is needed to determine its contribution to clinical as-
complete motor and sensory deficits affecting head, trunk, and limbs sessment of patients with GBS and its correlation with long-term
(locked-in syndrome), and manifest with quadriplegia, anarthria, dys- functional outcomes.
phagia, horizontal gaze palsy with or without preservation of vertical T-tau is a microtubule-
associated protein expressed in both
gaze, vertigo, and altered level of consciousness, typically with hy- central and peripheral axons. Plasma but not CSF levels of T-tau
peracute onset. Patients with brainstem strokes have hypertonia and are higher in GBS compared to disease-free and healthy controls,
hyperreflexia, unusual in peripheral neuropathy. Rhombencephalitis and plasma and CSF levels of T-t au do not correlate [69], possibly
can resemble GBS, but patients will have neighbourhood verte- suggesting that T-t au found in the blood of GBS patients originates
brobasilar signs, absent in GBS. Finally, Wernicke's encephalopathy from damaged distal peripheral nerve axons. No data are currently
(WE), manifesting with the triad ataxia, eye movement abnormalities available on the correlation of T-t au with I-RODS or other outcome
(ophthalmoplegia and/or nystagmus), and encephalopathy, can re- measures, and its potential role as a diagnostic, monitoring, or prog-
semble MFS or BBE. However, patients with MFS do not have altered nostic biomarker remains to be determined.
mental status or higher cortical dysfunction, and are areflexic. BBE
patients may develop a similar encephalopathy but, typically, have
pathologically brisk deep tendon reflexes. TR E ATM E NT
Current disease-
m odifying therapies in GBS include IVIg and
O U TCO M E M E A S U R E S I N G B S PLEX. Corticosteroids are not recommended in GBS: intra-
venous methylprednisolone (IVMP) does not improve neuro-
Outcome measures in the inflammatory neuropathies are usu- logical outcome [70, 71] and IVMP in combination with IVIg is
ally ordinal composite measures, and sum scores are generally not beneficial [72]. Oral corticosteroids may delay recovery or
used. In GBS, disease activity and clinical disability are measured cause side effects [70, 73]. Finally, a less common treatment is
|
14681331, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ene.16365 by Cochrane Mexico, Wiley Online Library on [07/06/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
10 of 15 BELLANTI and RINALDI
immunoabsorption (IA), which involves selectively removing IgG infusion of plasma or human albumin solution. A further group of
from the blood by passing separated plasma through an absorp- complications pertains to the vascular access, including line infec-
tion column. Although case reports and small case series suggest tion, sepsis, haematomas, venous thrombosis, and vascular damage
that IA may be safe and effective, it is expensive and not avail- [76].
able in all countries, and currently not recommended as first-or
second-line therapy for GBS [2].
IVIg and PLEX regimens
Whom and when to treat The usual IVIg regimen for GBS is 2 g/kg given over 5 days. This is
associated with a lower relapse (or treatment-related fluctuation)
If patients with GBS are to be treated with disease-modifying ther- risk compared to 2 g/kg over 2 days only [2]. Patients with severe
apy, then this should be started as early as possible within their dis- GBS and poor prognosis should receive one course of IVIg and do
ease course. IVIg and PLEX are thought to work by disrupting the not benefit from a second immunoglobulin dose, which increases
disimmune processes driving nerve damage, preventing further the risk of side effects (thromboembolic and vascular) without im-
nerve injury, and facilitating functional recovery. At nadir or dur- proving neurological outcome [77, 78]. In GBS patients who can
ing the recovery phase, when neuropathic processes have resolved, still walk unaided but are unable to run, two plasma exchanges
such treatments will no longer be of benefit. Moreover, if further within the first 2 weeks of disease onset are recommended. Four
nerve injury can be prevented before it occurs, and/or conduction to five plasma exchanges over 1 or 2 weeks, with a total exchanged
block reversed before axonal degeneration ensues, then the extent volume of 12–15 L, are recommended in patients who are still am-
of disability at nadir is likely to be reduced and the speed of recovery bulatory but have evidence of rapid deterioration (dyspnoea, bul-
increased. Trial evidence has only shown benefit with IVIg started bar, or autonomic dysfunction). There is currently no evidence for
within 2 weeks, and PLEX started within 4 weeks, of the onset of any form of combination therapy, such as PLEX after IVIg or vice
weakness. However, it is generally felt that IVIg is also likely to be ef- versa.
fective up to 4 weeks. Conversely, patients with mild disease (or pure
MFS without GBS overlap) who are destined for a benign disease
course without treatment are unlikely to benefit from such therapies PRO G N OS TI C CO N S I D E R ATI O N S
even within these timeframes and may be harmed by them. Figure 5
illustrates a decision-making algorithm for whether and when to GBS is a treatable disease and most patients ultimately recover.
treat based on timing and clinical features. About 80% of patients will walk independently and over 50% re-
turn to their previous baseline within a year [79]. However, up to
10% of patients require several months of mechanical ventilation,
IVIg versus PLEX and more than 10% are left with severe disability. Approximately
20% of patients who become ventilator-dependent and 3–7% of all
Head-to-head randomised trials directly comparing IVIg with PLEX GBS patients die [79–81], usually due to sepsis, acute respiratory
have shown no significant difference in treatment efficacy, but IVIg distress syndrome, pulmonary embolism, or cardiac arrest [82].
is less likely to be discontinued than PLEX [74]. The choice of which Negative prognostic factors include age >60 years, rapid onset of
therapy to offer is mostly based on pragmatic considerations and weakness (less than 7 days from symptom onset to hospital admis-
patient/clinician preference, and the latest guidelines do not support sion), severe weakness on admission, the need for mechanical ven-
one treatment over the other [2]. Plasma exchange is not available in tilation, preceding diarrhoea, and features of severe neuropathy on
all centres, and IVIg might be logistically easier. IVIg is generally well electrophysiology.
tolerated but may cause thromboembolism, especially in patients It should be noted that death and disability rates in GBS vary
with a previous thrombotic event [75]. PLEX is also well tolerated significantly between developed and developing countries, largely
but can cause hypotension from rapid fluid shifts, fluid overload, and because treatments like IVIg and PLEX are often unavailable.
vasovagal episodes. Such complications may be more frequent in Additionally, inadequate rehabilitation services further contribute to
GBS patients who often have labile blood pressure due to autonomic disparities in functional outcomes. Small volume plasma exchange
nervous system involvement. Clinicians should consider suspending (SVPE), the repeated removal of small volumes of plasma over several
anti-hypertensive therapy on treatment days. Allergic or anaphylac- days via sedimentation of patient whole blood, has been evaluated
tic reactions are rare and, when occurring, usually result from the as a potential low-cost alternative treatment [83]. Although SVPE
F I G U R E 5 Guillain-Barré syndrome (GBS) treatment flowchart. *Modified Erasmus GBS Respiratory Insufficiency Score (mEGRIS) ≥3/
modified Erasmus GBS Outcome Score (mEGOS) ≥4. A-CIDP, acute-onset chronic inflammatory demyelinating polyradiculoneuropathy; AIN,
autoimmune nodopathy; IVIg, intravenous immunoglobulin; PLEX, plasma exchange.
14681331, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ene.16365 by Cochrane Mexico, Wiley Online Library on [07/06/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
|11 of 15
GUILLAIN-BARRÉ SYNDROME: A REVIEW
|
14681331, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ene.16365 by Cochrane Mexico, Wiley Online Library on [07/06/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
12 of 15 BELLANTI and RINALDI
appears feasible and safe, randomised control trials are needed to IgG degradation
determine its clinical efficacy.
Imlifidase is an IgG-degrading enzyme derived from Streptococcus
pyogenes which can cleave and inactivate all four subclasses of
Prediction of respiratory insufficiency human IgG. IgG metabolising enzymes are able to block complement
activation in vitro [91] and facilitate recovery in rabbit models of
Early recognition of severe GBS is pivotal to enable prompt interven- GBS with axonal injury, where they also reduce axonal degeneration
tion. The Erasmus GBS Respiratory Insufficiency Score (EGRIS) is a [92]. In light of these findings, imlifidase is currently being evaluated
clinical model used to predict the probability of developing respira- in an open-label, multicentre, phase II study as potential treatment
tory failure within the first week of admission in patients with GBS. for GBS.
The estimated risk is based on the presence or absence of specific
clinical features upon hospital admission [84]. The modified EGRIS
(mEGRIS), the latest version of the model, can be used to predict CO N C LU S I O N S
respiratory failure at any point during the first 2 months from dis-
ease onset, not just in the first week. On a practical level, such Guillain-Barré syndrome is a postinfectious, immune-mediated pe-
risk is greater in patients with rapid disease progression in the first ripheral neuropathy. As our knowledge continues to expand, its
4 weeks, bulbar palsy, and weakness of neck flexion and hip flexion. pathophysiological mechanisms remain partially understood and
These patients should be monitored closely, with a low threshold for many questions are yet to be answered. The focus of care should
admission to the intensive care unit (ICU) if required [85, 86]. remain early diagnosis and treatment, to prevent severe axonal dam-
age and minimise long-term disability. Meanwhile, novel therapies
and neuropathy fluid biomarkers are under ongoing evaluation and
Prediction of functional outcome may improve clinical management in the foreseeable future.
New potential therapies for GBS are currently under evaluation but REFERENCES
none are yet routinely used in clinical practice. Clinical trials of ecu- 1. Guillain G. Sur un syndrome de radiculo- nevrite avec hyperal-
lizumab, a humanised monoclonal antibody against the complement buminose du liquide cephalo- rachidien sans reaction cellulaire.
Remarques sur les caracters cliniques et graphiques des reflexes
protein C5, have so far not provided evidence of efficacy in patients
tendineux. Bull Mem Soc Med Hop Paris. 1916;40:1462-1470.
with severe GBS, either due to failure to meet the primary endpoint 2. van Doorn PA, Van den Bergh PYK, Hadden RDM, et al. European
[88] or premature interruption [89]. At present, given lack of defini- Academy of Neurology/Peripheral Nerve Society guideline on di-
tively proven efficacy, together with high costs and potential side agnosis and treatment of Guillain–Barré syndrome. Eur J Neurol.
2023;30:3646-3674. doi:10.1111/ene.16073
effects, eculizumab is not recommended for the treatment of GBS,
3. Sejvar JJ, Baughman AL, Wise M, Morgan OW. Population incidence
and large, randomised controlled trials are needed to establish its of Guillain-Barré syndrome: a systematic review and meta-analysis.
potential clinical utility. Inhibition of C1q factor with the monoclonal Neuroepidemiology. 2011;36(2):123-133. doi:10.1159/000324710
antibody ANX005 in GBS has shown preliminary evidence of effi- 4. Matsui N, Nodera H, Kuzume D, et al. Guillain-Barré syndrome
in a local area in Japan, 2006–2015: an epidemiological and clin-
cacy (NfL reduction and improvement of GBS disability score in the
ical study of 108 patients. Eur J Neurol. 2018;25(5):718-724.
first weeks after treatment) [90] and remains under evaluation. doi:10.1111/ene.13569
|
14681331, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ene.16365 by Cochrane Mexico, Wiley Online Library on [07/06/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GUILLAIN-BARRÉ SYNDROME: A REVIEW 13 of 15
5. Cheng Q, Wang DS, Jiang GX, et al. Distinct pattern of age-specific States. JAMA Netw Open. 2023;6(2):e2253845. doi:10.1001/
incidence of Guillain-Barré syndrome in Harbin, China. J Neurol. jamanetworkopen.2022.53845
2002;249(1):25-32. doi:10.1007/pl00007844 24. Keh RYS, Scanlon S, Datta- Nemdharry P, et al. COVID- 19 vac-
6. Howlett WP, Vedeler CA, Nyland H, Aarli JA. Guillain-Barré syn- cination and Guillain- Barré syndrome: analyses using the
drome in northern Tanzania: a comparison of epidemiological National Immunoglobulin Database. Brain. 2023;146(2):739-748.
and clinical findings with western Norway. Acta Neurol Scand. doi:10.1093/brain/awac067
1996;93(1):44-49. doi:10.1111/j.1600-0 404.1996.tb00169.x 25. Patone M, Handunnetthi L, Saatci D, et al. Neurological com-
7. Sipilä JOT, Soilu-Hänninen M, Ruuskanen JO, Rautava P, Kytö V. plications after first dose of COVID- 19 vaccines and SARS-
Epidemiology of Guillain-Barré syndrome in Finland 2004–2014. J CoV-2 infection. Nat Med. 2021;27(12):2144-2153. doi:10.1038/
Peripher Nerv Syst. 2017;22:440-445. s41591-021-01556-7
8. Rivera-Lillo G, Torres- C astro R, Burgos PI, et al. Incidence of 26. Gensicke H, Datta AN, Dill P, Schindler C, Fischer D. Increased
Guillain-
Barré syndrome in Chile: a population- based study. J incidence of Guillain-Barré syndrome after surgery. Eur J Neurol.
Peripher Nerv Syst. 2016;21(4):339-344. doi:10.1111/jns.12182 2012;19(9):1239-1244. doi:10.1111/j.1468-1331.2012.03730.x
9. Islam Z, Jacobs BC, Islam MB, Mohammad QD, Diorditsa S, 27. Rudant J, Dupont A, Mikaeloff Y, Bolgert F, Coste J, Weill A. Surgery
Endtz HP. High incidence of Guillain-Barré Syndrome in children, and risk of Guillain-Barré syndrome: a French nationwide epidemi-
Bangladesh. Emerg Infect Dis. 2011;17:1317–1318. ologic study. Neurology. 2018;91(13):e1220-e1227. doi:10.1212/
10. Webb AJ, Brain SA, Wood R, Rinaldi S, Turner MR. Seasonal varia- WNL.0000000000006246
tion in Guillain-Barré syndrome: a systematic review, meta-analysis 28. Hocker S, Nagarajan E, Rubin M, Wijdicks EFM. Clinical factors asso-
and Oxfordshire cohort study. J Neurol Neurosurg Psychiatry ciated with Guillain-Barré syndrome following surgery. Neurol Clin
2015;86:1196–1201. Pract. 2018;8(3):201-206. doi:10.1212/CPJ.0000000000000451
11. Jackson BR, Zegarra JA, López-Gatell H, et al. Binational outbreak of 29. Zhang L, Arrington S, Keung YK. Guillain- Barré syndrome
Guillain-Barré syndrome associated with Campylobacter jejuni in- after transplantation. Leuk Lymphoma. 2008;49(2):291-297.
fection, Mexico and USA, 2011. Epidemiol Infect. 2014;142(5):1089- doi:10.1080/10428190701760003
1099. doi:10.1017/S0950268813001908 30. Gu Y, Menzies AM, Long GV, Fernando SL, Herkes G. Immune
12. Styczynski AR, Malta JMAS, Krow-Lucal ER, et al. Increased rates mediated neuropathy following checkpoint immunotherapy. J Clin
of Guillain-Barré syndrome associated with Zika virus outbreak Neurosci. 2017;45:14-17. doi:10.1016/j.jocn.2017.07.014
in the Salvador metropolitan area, Brazil. PLoS Negl Trop Dis. 31. Haymaker W, Kebnohan JW. The Landry-Guillain-Barré syndrome:
2017;11(8):e0005869. doi:10.1371/journal.pntd.0005869 a clinicopathologic report of fifty fatal cases and a critique of the
13. Shahrizaila N, Lehmann HC, Kuwabara S. Guillain- Barré syn- literature. Medicine. 1949;28(1):59.
drome. Lancet. 2021;397(10280):1214-1228. doi:10.1016/ 32. Súkeníková L, Mallone A, Schreiner B, Ripellino P, Nilsson J, Stoffel
S0140-6736(21)00517-1 M, Ulbrich SE, Sallusto F, Latorre D. Autoreactive T cells target pe-
14. Willison HJ, Jacobs BC, Van Doorn PA. Guillain- Barré syn- ripheral nerves in Guillain–Barré syndrome. Nature 2024;626:160–
drome. Lancet. 2016;388(10045):717-727. doi:10.1016/ 168. doi:10.1038/s41586-023-06916-6,
S0140-6736(16)00339-1 33. Griffin JW, Li CY, Macko C, et al. Early nodal changes in the acute
15. Doets AY, Verboon C, van den Berg B, et al. Regional variation motor axonal neuropathy pattern of the Guillain-Barré syndrome. J
of Guillain- Barré syndrome. Brain. 2018;141(10):2866-2877. Neurocytol. 1996;25(1):33-51. doi:10.1007/BF02284784
doi:10.1093/brain/awy232 34. Hafer-Macko C, Hsieh ST, Ho TW, et al. Acute motor axonal neu-
16. Meyer Sauteur PM, Huizinga R, Tio-G illen AP, et al. Mycoplasma ropathy: an antibody-mediated attack on axolemma. Ann Neurol.
pneumoniae triggering the Guillain- B arré syndrome: a case- 1996;40(4):635-644. doi:10.1002/ana.410400414
control study. Ann Neurol. 2016;80(4):566-580. doi:10.1002/ 35. McGonigal R, Rowan EG, Greenshields KN, et al. Anti-GD1a anti-
ana.24755 bodies activate complement and calpain to injure distal motor nodes
17. Kusunoki S, Shiina M, Kanazawa I. Anti-Gal-C antibodies in GBS of Ranvier in mice. Brain. 2010;133(7):1944-1960. doi:10.1093/
subsequent to mycoplasma infection: evidence of molecular mim- brain/awq119
icry. Neurology. 2001;57(4):736-738. doi:10.1212/wnl.57.4.736 36. Susuki K, Rasband MN, Tohyama K, et al. Anti-GM1 antibodies
18. Langmuir AD, Bregman DJ, Kurland LT, Nathanson N, Victor M. An cause complement-mediated disruption of sodium channel clusters
epidemiologic and clinical evaluation of Guillain-Barré syndrome in peripheral motor nerve fibers. J Neurosci. 2007;27(15):3956-
reported in association with the administration of swine influenza 3967. doi:10.1523/JNEUROSCI.4401-06.2007
vaccines. Am J Epidemiol. 1984;119(6):841-879. doi:10.1093/ox- 37. Halstead SK, Zitman FMP, Humphreys PD, et al. Eculizumab pre-
fordjournals.aje.a113809 vents anti-g anglioside antibody-m ediated neuropathy in a mu-
19. Salmon DA, Dudley MZ, Carleton BC. Guillain-Barré syndrome fol- rine model. Brain. 2008;131(5):1197-1208. doi:10.1093/brain/
lowing influenza vaccines affords opportunity to improve vaccine awm316
confidence. J Infect Dis. 2021;223(3):355-358. doi:10.1093/infdis/ 38. McGonigal R, Cunningham ME, Smyth D, et al. The endogenous
jiaa544 calpain inhibitor calpastatin attenuates axon degeneration in mu-
20. Lehmann HC, Hartung HP, Kieseier BC, Hughes RAC. Guillain- rine Guillain-Barré syndrome. J Peripher Nerv Syst. 2023;28:4-16.
Barré syndrome after exposure to influenza virus. Lancet Infect Dis. doi:10.1111/jns.12520
2010;10(9):643-651. doi:10.1016/S1473-3 099(10)70140-7 39. Halstead SK, O'Hanlon GM, Humphreys PD, et al. Anti-disialoside
21. Salmon DA, Proschan M, Forshee R, et al. Association between antibodies kill perisynaptic Schwann cells and damage motor nerve
Guillain-Barré syndrome and influenza A (H1N1) 2009 monova- terminals via membrane attack complex in a murine model of neu-
lent inactivated vaccines in the USA: a meta- analysis. Lancet. ropathy. Brain. 2004;127(Pt 9):2109-2123. doi:10.1093/brain/
2013;381(9876):1461-1468. doi:10.1016/S0140-6736(12)62189-8 awh231
22. Goud R, Lufkin B, Duffy J, et al. Risk of Guillain-Barré syndrome 40. McGonigal R, Campbell CI, Barrie JA, et al. Schwann cell nodal
following recombinant zoster vaccine in Medicare beneficia- membrane disruption triggers bystander axonal degenera-
ries. JAMA Intern Med. 2021;181(12):1623-1630. doi:10.1001/ tion in a Guillain- Barré syndrome mouse model. J Clin Invest.
jamainternmed.2021.6227 2022;132(14):e158524. doi:10.1172/JCI158524
23. Abara WE, Gee J, Marquez P, et al. Reports of Guillain- 41. Campbell CI, McGonigal R, Barrie JA, et al. Complement inhibition
Barré syndrome after COVID- 19 vaccination in the United prevents glial nodal membrane injury in a GM1 antibody-mediated
|
14681331, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ene.16365 by Cochrane Mexico, Wiley Online Library on [07/06/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
14 of 15 BELLANTI and RINALDI
mouse model. Brain Commun. 2022;4(6):fcac306. doi:10.1093/ 59. Howard RS. Poliomyelitis and the postpolio syndrome. BMJ.
braincomms/fcac306 2005;330(7503):1314-1318.
42. Kichi K, Morita D, Kanzaki M, et al. Ganglioside complexes as 60. Wakerley BR, Yuki N. Mimics and chameleons in Guillain–Barré
new target antigens in Guillain- Barré syndrome. Ann Neurol. and Miller Fisher syndromes. Pract Neurol. 2015;15(2):90-99.
2004;56(4):567-571. doi:10.1002/ana.20222 doi:10.1136/practneurol-2014-0 00937
43. Rinaldi S, Brennan KM, Kalna G, et al. Antibodies to heteromeric 61. Solomon T, Lewthwaite P, Perera D, Cardosa MJ, McMinn P, Ooi
glycolipid complexes in Guillain- Barré syndrome. PLoS One. MH. Virology, epidemiology, pathogenesis, and control of en-
2013;8(12):e82337. doi:10.1371/journal.pone.0082337 terovirus 71. Lancet Infect Dis. 2010;10(11):778-790. doi:10.1016/
44. Wu LY, Zhou Y, Qin C, Hu BL. The effect of TNF-alpha, FcγR and S1473-3 099(10)70194-8
CD1 polymorphisms on Guillain-Barré syndrome risk: evidences 62. Kranick SM, Nath A. Neurologic complications of HIV- 1 in-
from a meta- analysis. J Neuroimmunol. 2012;243(1–2):18-24. fection and its treatment in the era of antiretroviral ther-
doi:10.1016/j.jneuroim.2011.12.003 apy. Continuum. 2012;18(6):1319-1337. doi:10.1212/01.
45. Geleijns K, Roos A, Houwing- Duistermaat JJ, et al. Mannose- CON.0000423849.24900.ec
binding lectin contributes to the severity of Guillain- Barré 63. Gandhi Mehta RK, Caress JB, Rudnick SR, Bonkovsky HL. Porphyric
syndrome. J Immunol. 2006;177(6):4211-4217. doi:10.4049/ neuropathy. Pract Neurol. 2021;64(2):140-152. doi:10.1002/
jimmunol.177.6.4211 mus.27232
46. Leonhard SE, Mandarakas MR, Gondim FAA, et al. Diagnosis and 64. Nasir M, Obrocki R, Krommyda M, Malek N. Conus medullaris syn-
management of Guillain- Barré syndrome in ten steps. Nat Rev drome as a presenting feature of MOG-associated disease. Pract
Neurol. 2019;15(11):671-683. doi:10.1038/s41582-019-0250-9 Neurol. 2023;23(4):314-316. doi:10.1136/pn-2022-0 03560
47. Fokke C, van den Berg B, Drenthen J, Walgaard C, van Doorn PA, 65. Tri SL, Vinh KN, Dang TQ, Umapathi T. Myoedema: a forgotten sign
Jacobs BC. Diagnosis of Guillain-Barré syndrome and validation of in acute colchicine myopathy. BMJ Case Rep. 2023;16(10):e257076.
Brighton criteria. Brain. 2014;137(Pt 1):33-43. doi:10.1093/brain/ doi:10.1136/bcr-2023-257076
awt285 66. Martín-A guilar L, Camps-Renom P, Lleixà C, et al. Serum neuro-
48. Bellanti R, Keddie S, Lunn MP, Rinaldi S. Ultrasensitive assay filament light chain predicts long-term prognosis in Guillain-Barré
technology and fluid biomarkers for the evaluation of peripheral syndrome patients. J Neurol Neurosurg Psychiatry. 2021;92(1):70-77.
nerve disease. J Neurol Neurosurg Psychiatry. 2024;95:114-124. doi:10.1136/jnnp-2020-323899
doi:10.1136/jnnp-2023-332031 67. Petzold A. The 2022 Lady Estelle Wolfson lectureship on neurofil-
49. Pestronk A, Choksi R, Yee WC, Kornberg AJ, Lopate G, Trotter aments. J Neurochem. 2022;163(3):179-219. doi:10.1111/jnc.15682
J. Serum antibodies to heparan sulfate glycosaminoglycans in 68. Berciano J. Peripherin is a biomarker of axonal damage in
Guillain-
Barré syndrome and other demyelinating polyneurop- Guillain-Barré syndrome: a pathophysiological annotation. Brain.
athies. J Neuroimmunol. 1998;91(1–2):204-209. doi:10.1016/ 2024;147:e1-e2. doi:10.1093/brain/awad277
s0165-5728(98)00182-9 69. Kmezic I, Samuelsson K, Finn A, et al. Neurofilament light chain
50. Vucic S, Cairns KD, Black KR, Chong PST, Cros D. Neurophysiologic and total tau in the differential diagnosis and prognostic evaluation
findings in early acute inflammatory demyelinating polyradic- of acute and chronic inflammatory polyneuropathies. Eur J Neurol.
uloneuropathy. Clin Neurophysiol. 2004;115(10):2329-2335. 2022;29(9):2810-2822. doi:10.1111/ene.15428
doi:10.1016/j.clinph.2004.05.009 70. Hughes RA, Brassington R, Gunn AA, van Doorn PA.
51. Scarpino M, Lolli F, Carrai R, et al. Diagnostic accuracy of neu- Corticosteroids for Guillain- B arré syndrome. Cochrane
rophysiological criteria for early diagnosis of AIDP: a prospec- Database Syst Rev. 2016;10:CD001446. doi:10.1002/14651858.
tive study. Neurophysiol Clin. 2016;46(1):35-42. doi:10.1016/j. CD001446.pub5
neucli.2015.12.008 71. Group GBSST. Double-blind trial of intravenous methylpredniso-
52. Chanson JB, Echaniz- L aguna A. Early electrodiagnostic ab- lone in Guillain-Barré syndrome. Lancet. 1993;341(8845):586-590.
normalities in acute inflammatory demyelinating polyneurop- doi:10.1016/0140-6736(93)90351-G
athy: a retrospective study of 58 patients. Clin Neurophysiol. 72. van Koningsveld R, Schmitz PI, Meché FG, et al. Effect of methyl-
2014;125(9):1900-1905. doi:10.1016/j.clinph.2014.01.007 prednisolone when added to standard treatment with intravenous
53. Umapathi T, Lim CSJ, Ng BCJ, Goh EJH, Ohnmar O. A simplified, immunoglobulin for Guillain- Barré syndrome: randomised trial.
graded, electrodiagnostic criterion for Guillain- Barré Syndrome Lancet. 2004;363:192-196.
that incorporates sensory nerve conduction studies. Sci Rep. 73. Hughes RA, Newsom- Davis JM, Perkin GD, Pierce JM.
2019;9:7724. Controlled trial prednisolone in acute polyneuropathy. Lancet.
54. Derksen A, Ritter C, Athar P, et al. Sural sparing pattern discrim- 1978;2(8093):750-753. doi:10.1016/s0140-6736(78)92644-2
inates Guillain- Barré syndrome from its mimics. Muscle Nerve. 74. Hughes RAC, Swan AV, van Doorn PA. Intravenous immunoglob-
2014;50(5):780-784. doi:10.1002/mus.24226 ulin for Guillain- Barré syndrome. Cochrane Database Syst Rev.
55. Roux T, Debs R, Maisonobe T, et al. Rituximab in chronic inflam- 2014;2014(9):CD002063. doi:10.1002/14651858.CD002063.pub6
matory demyelinating polyradiculoneuropathy with associated 75. Kapoor M, Hunt I, Spillane J, et al. IVIg-exposure and thromboem-
diseases. J Peripher Nerv Syst. 2018;23(4):235-240. doi:10.1111/ bolic event risk: findings from the UK Biobank. J Neurol Neurosurg
jns.12287 Psychiatry. 2022;93(8):876-885. doi:10.1136/jnnp-2022-328881
56. Burnor E, Yang L, Zhou H, et al. Neurofascin antibodies in au- 76. Fehmi J, Bellanti R, Misbah SA, Bhattacharjee A, Rinaldi S.
toimmune, genetic, and idiopathic neuropathies. Neurology. Treatment of CIDP. Pract Neurol 2023;23:46–53. doi:10.1136/
2018;90(1):e31-e38. doi:10.1212/WNL.0000000000004773 pn-2021-0 02991
57. Querol L, Rojas- García R, Diaz- Manera J, et al. Rituximab in 77. Walgaard C, Jacobs BC, Lingsma HF, et al. Second intravenous im-
treatment- resistant CIDP with antibodies against paranodal munoglobulin dose in patients with Guillain-Barré syndrome with
proteins. Neurol Neuroimmunol Neuroinflamm. 2015;2(5):e149. poor prognosis (SID-GBS): a double-blind, randomised, placebo-
doi:10.1212/NXI.0000000000000149 controlled trial. Lancet Neurol. 2021;20(4):275-283. doi:10.1016/
58. Ruts L, van Koningsveld R, Jacobs BC, van Doorn PA. Determination S1474-4 422(20)30494-4
of pain and response to methylprednisolone in Guillain- Barré 78. Lunn MP. Preventing expensive harms in Guillain- Barré syn-
syndrome. J Neurol. 2007;254(10):1318-1322. doi:10.1007/ drome. Lancet Neurol. 2021;20(4):249-251. doi:10.1016/
s00415- 0 06- 0515-2 S1474-4 422(21)00041-7
|
14681331, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ene.16365 by Cochrane Mexico, Wiley Online Library on [07/06/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GUILLAIN-BARRÉ SYNDROME: A REVIEW 15 of 15
79. Rajabally YA, Uncini A. Outcome and its predictors in Guillain- 91. Takahashi R, Yuki N. Streptococcal IdeS: therapeutic potential for
Barre syndrome. J Neurol Neurosurg Psychiatry. 2012;83(7):711-718. Guillain–Barré syndrome. Sci Rep. 2015;5:10809. doi:10.1038/
doi:10.1136/jnnp-2011-3 01882 srep10809
80. Chiò A, Cocito D, Leone M, et al. Guillain-Barré syndrome: a prospec- 92. Wang Y, Shi Q, Lv H, et al. IgG-degrading enzyme of Streptococcus
tive, population- based incidence and outcome survey. Neurology. pyogenes (IdeS) prevents disease progression and facilitates im-
2003;60(7):1146-1150. doi:10.1212/01.wnl.0000055091.96905.d0 provement in a rabbit model of Guillain- Barré syndrome. Exp
81. van den Berg B, Bunschoten C, van Doorn PA, Jacobs BC. Mortality Neurol. 2017;291:134-140. doi:10.1016/j.expneurol.2017.02.010
in Guillain- Barre syndrome. Neurology. 2013;80(18):1650-1654. 93. Lee SH, Lim GH, Kim JS, et al. Acute ophthalmoplegia (with-
doi:10.1212/WNL.0b013e3182904fcc out ataxia) associated with anti- GQ1b antibody. Neurology.
82. Lawn ND, Wijdicks EF. Fatal Guillain-Barré syndrome. Neurology. 2008;71:426-429. doi:10.1212/01.wnl.0000324266.95814.74
1999;52(3):635-638. doi:10.1212/wnl.52.3.635 94. Thomma RCM, Fokke C, Walgaard C, et al. High and persistent an-
83. Islam MB, Islam Z, Rahman S, et al. Small volume plasma exchange ti-GM1 antibody titers are associated with poor clinical recovery
for Guillain-Barré syndrome in resource poor settings: a safety and in Guillain-
Barré syndrome. Neurol Neuroimmunol Neuroinflamm.
feasibility study. Pilot Feasibility Stud. 2017;3(1):40. doi:10.1186/ 2023;10(4):e200107. doi:10.1212/NXI.0000000000200107
s40814-017-0185-0 95. Kim JK, Bae JS, Kim DS, et al. Prevalence of anti-ganglioside anti-
84. Doets AY, Walgaard C, Lingsma HF, et al. International validation bodies and their clinical correlates with Guillain-Barré syndrome in
of the Erasmus Guillain-Barré Syndrome respiratory insufficiency Korea: a nationwide multicenter study. J Clin Neurol. 2014;10(2):94-
score. Ann Neurol. 2022;91:521-531. 100. doi:10.3988/jcn.2014.10.2.94
85. Walgaard C, Lingsma HF, Ruts L, et al. Prediction of respiratory 96. Yamagishi Y, Kuwahara M, Suzuki H, et al. Serum IgG anti-GD1a an-
insufficiency in Guillain-Barré syndrome. Ann Neurol. 2010;67:781- tibody and mEGOS predict outcome in Guillain-Barré syndrome. J
787. doi:10.1002/ana.21976 Neurol Neurosurg Psychiatry. 2020;91(12):1339-1342. doi:10.1136/
86. Luijten LWG, Doets AY, Arends S, et al. Modified Erasmus GBS jnnp-2020-323960
respiratory insufficiency score: a simplified clinical tool to predict 97. Koga M, Yuki N, Ariga T, Morimatsu M, Hirata K. Is IgG anti-GT1a
the risk of mechanical ventilation in Guillain-Barré syndrome. J antibody associated with pharyngeal-cervical-brachial weakness or
Neurol Neurosurg Psychiatry. 2023;94(4):300-308. doi:10.1136/ oropharyngeal palsy in Guillain-Barré syndrome? J Neuroimmunol.
jnnp-2022-329937 1998;86(1):74-79. doi:10.1016/s0165-5728(98)00016-2
87. Walgaard C, Lingsma HF, Ruts L, van Doorn PA, Steyerberg
EW, Jacobs BC. Early recognition of poor prognosis in Guillain-
Barre syndrome. Neurology. 2011;76(11):968-975. doi:10.1212/ S U P P O R T I N G I N FO R M AT I O N
WNL.0b013e3182104407 Additional supporting information can be found online in the
88. Misawa S, Kuwabara S, Sato Y, et al. Safety and efficacy of ecu-
Supporting Information section at the end of this article.
lizumab in Guillain-Barré syndrome: a multicentre, double-blind,
randomised phase 2 trial. Lancet Neurol. 2018;17(6):519-529.
doi:10.1016/S1474-4 422(18)30114-5
89. Davidson AI, Halstead SK, Goodfellow JA, et al. Inhibition of com-
plement in Guillain-Barré syndrome: the ICA-GBS study. J Peripher How to cite this article: Bellanti R, Rinaldi S. Guillain-Barré
Nerv Syst. 2017;22(1):4-12. doi:10.1111/jns.12194 syndrome: A comprehensive review. Eur J Neurol.
90. Annexon Biosciences. Annexon Biosciences reports top-line Phase 2024;00:e16365. doi:10.1111/ene.16365
1b results for novel C1q inhibitor ANX005 in Guillain-Barré syn-
drome. Accessed January 11, 2024. https://annexonbio.com/news/
annexon-biosciences-repor t s-top-line-phase-1b-result s-for-novel
-c1q-inhibitor-anx005-in-guillain-barre-syndrome/